Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Am Chem Soc ; 144(28): 12893-12900, 2022 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-35786910

RESUMO

Bioorthogonal catalysis using transition-metal catalysts (TMCs) provides a toolkit for the in situ generation of imaging and therapeutic agents in biological environments. Integrating TMCs with nanomaterials mimics key properties of natural enzymes, providing bioorthogonal "nanozymes". ZnS nanoparticles provide a platform for bioorthogonal nanozymes using ruthenium catalysts embedded in self-assembled monolayers on the particle surface. These nanozymes uncage allylated profluorophores and prodrugs. The ZnS core combines the non-toxicity and degradability with the enhancement of Ru catalysis through the release of thiolate surface ligands that accelerate the rate-determining step in the Ru-mediated deallylation catalytic cycle. The maximum rate of reaction (Vmax) increases ∼2.5-fold as compared to the non-degradable gold nanoparticle analogue. The therapeutic potential of these bioorthogonal nanozymes is demonstrated by activating a chemotherapy drug from an inactive prodrug with efficient killing of cancer cells.


Assuntos
Nanopartículas Metálicas , Pró-Fármacos , Rutênio , Elementos de Transição , Catálise , Ouro , Pró-Fármacos/farmacologia , Sulfetos , Compostos de Zinco
2.
Pharm Res ; 39(6): 1197-1204, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35297498

RESUMO

PURPOSE: Cytosolic delivery of proteins accesses intracellular targets for chemotherapy and immunomodulation. Current delivery systems utilize inefficient endosomal pathways of uptake and escape that lead to degradation of delivered cargo. Cationic poly(oxanorbornene)imide (PONI) polymers enable highly efficient cytosolic delivery of co-engineered proteins, but aggregation and denaturation in solution limits shelf life. In the present study we evaluate polymer-protein nanocomposite vehicles as candidates for lyophilization and point-of-care resuspension to provide a transferrable technology for cytosolic protein delivery. METHODS: Self-assembled nanocomposites of engineered poly(glutamate)-tagged (E-tagged) proteins and guanidinium-functionalized PONI homopolymers were generated, lyophilized, and stored for 2 weeks. After reconstitution and delivery, cytosolic access of E-tagged GFP cargo (GFPE15) was assessed through diffuse cytosolic and nuclear fluorescence, and cell killing with chemotherapeutic enzyme Granzyme A (GrAE10). Efficiency was quantified between freshly prepared and lyophilized samples. RESULTS: Reconstituted nanocomposites retained key structural features of freshly prepared assemblies, with minimal loss of material. Cytosolic delivery (> 80% efficiency of freshly prepared nanocomposites) of GFPE15 was validated in several cell lines, with intracellular access validated and quantified through diffusion into the nucleus. Delivery of GrAE10 elicited significant tumorigenic cell death. Intracellular access of cytotoxic protein was validated through cell viability. CONCLUSION: Reconstituted nanocomposites achieved efficient cytosolic delivery of protein cargo and demonstrated therapeutic applicability with delivery of GrAE10. Overall, this strategy represents a versatile and highly translatable method for cytosolic delivery of proteins.


Assuntos
Polímeros , Proteínas , Citosol/metabolismo , Endossomos/metabolismo , Liofilização , Polímeros/química , Proteínas/química
3.
J Am Chem Soc ; 143(12): 4758-4765, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33705125

RESUMO

Intracellular protein delivery enables selective regulation of cellular metabolism, signaling, and development through introduction of defined protein quantities into the cell. Most applications require that the delivered protein has access to the cytosol, either for protein activity or as a gateway to other organelles such as the nucleus. The vast majority of delivery vehicles employ an endosomal pathway however, and efficient release of entrapped protein cargo from the endosome remains a challenge. Recent research has made significant advances toward efficient cytosolic delivery of proteins using polymers, but the influence of polymer architecture on protein delivery is yet to be investigated. Here, we developed a family of dendronized polymers that enable systematic alterations of charge density and structure. We demonstrate that while modulation of surface functionality has a significant effect on overall delivery efficiency, the endosomal release rate can be highly regulated by manipulating polymer architecture. Notably, we show that large, multivalent structures cause slower sustained release, while rigid spherical structures result in rapid burst release.


Assuntos
Citosol/metabolismo , Polímeros/química , Engenharia de Proteínas , Proteínas/metabolismo , Animais , Linhagem Celular , Citosol/química , Humanos , Camundongos , Estrutura Molecular , Polímeros/metabolismo , Proteínas/química
4.
Bioconjug Chem ; 32(5): 891-896, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-33872490

RESUMO

Intracellular protein delivery is a transformative tool for biologics research and medicine. Delivery into the cytosol allows proteins to diffuse throughout the cell and access subcellular organelles. Inefficient delivery caused by endosomal entrapment is often misidentified as cytosolic delivery. This inaccuracy muddles what should be a key checkpoint in assessing delivery efficiency. Green fluorescent protein (GFP) is a robust cargo small enough to passively diffuse from the cytosol into the nucleus. Fluorescence of GFP in the nucleus is a direct readout for cytosolic access and effective delivery. Here, we highlight recent examples from the literature for the accurate assessment of cytosolic protein delivery using GFP fluorescence in the cytosol and nucleus.


Assuntos
Proteínas de Bactérias/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Proteínas Luminescentes/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Humanos
5.
J Am Chem Soc ; 142(9): 4349-4355, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32049533

RESUMO

Nanocarrier-mediated protein delivery is a promising strategy for fundamental research and therapeutic applications. However, the efficacy of the current platforms for delivery into cells is limited by endosomal entrapment of delivered protein cargo with concomitantly inefficient access to the cytosol and other organelles, including the nucleus. We report here a robust, versatile polymeric-protein nanocomposite (PPNC) platform capable of efficient (≥90%) delivery of proteins to the cytosol. We synthesized a library of guanidinium-functionalized poly(oxanorborneneimide) (PONI) homopolymers with varying molecular weights to stabilize and deliver engineered proteins featuring terminal oligoglutamate "E-tags". The polymers were screened for cytosolic delivery efficiency using imaging flow cytometry with cytosolic delivery validated using confocal microscopy and activity of the delivered proteins demonstrated through functional assays. These studies indicate that the PPNC platform provides highly effective and tunable cytosolic delivery over a wide range of formulations, making them robust agents for therapeutic protein delivery.


Assuntos
Portadores de Fármacos/metabolismo , Integrases/metabolismo , Proteínas Luminescentes/metabolismo , Ácido Poliglutâmico/metabolismo , Polímeros/metabolismo , Portadores de Fármacos/síntese química , Guanidinas/síntese química , Guanidinas/metabolismo , Células HEK293 , Células HeLa , Humanos , Imidas/síntese química , Imidas/metabolismo , Nanocompostos/química , Polímeros/síntese química , Engenharia de Proteínas , Proteína Vermelha Fluorescente
6.
Small ; 16(36): e2002084, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32347000

RESUMO

Industrial use of nanomaterials is rapidly increasing, making the effects of these materials on the environment and human health of critical concern. Standard nanotoxicity evaluation methods rely on detecting cell death or major dysfunction and will miss early signs of toxicity. In this work, the use of rapid and sensitive nanosensors that can efficiently detect subtle phenotypic changes on the cell surface following nanomaterial exposure is reported. Importantly, the method reveals significant phenotypic changes at dosages where other conventional methods show normal cellular activity. This approach holds promise in toxicological and pharmacological evaluations to ensure safer and better use of nanomaterials.


Assuntos
Técnicas Biossensoriais , Células , Nanopartículas , Toxicologia , Técnicas Biossensoriais/normas , Células/efeitos dos fármacos , Monitoramento Ambiental , Humanos , Nanopartículas/toxicidade , Toxicologia/instrumentação
7.
Chembiochem ; 21(19): 2759-2763, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32400081

RESUMO

We demonstrate here the protection of biorthogonal transition metal catalysts (TMCs) in biological environments by using self-assembled monolayers on gold nanoparticles (AuNPs). Encapsulation of TMCs in this hydrophobic environment preserves catalytic activity in presence of pH conditions and complex biological media that would deactivate free catalyst. Significantly, the protection affords by these nanozymes extends to isolation of the catalyst active site, as demonstrated by the independence of rate over a wide pH range, in strong contrast to the behavior of the free catalyst.


Assuntos
Complexos de Coordenação/química , Metais Pesados/química , Nanoestruturas/química , Catálise , Complexos de Coordenação/síntese química , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Metais Pesados/isolamento & purificação , Estrutura Molecular , Tamanho da Partícula
8.
Biol Pharm Bull ; 42(3): 304-311, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30828060

RESUMO

Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology has revolutionized therapeutic gene editing by providing researchers with a new method to study and cure diseases previously considered untreatable. While the full range and power of CRISPR technology for therapeutics is being elucidated through in vitro studies, translation to in vivo studies is slow. To date there is no totally effective delivery strategy to carry CRISPR components to the target site in vivo. The complexity of in vivo delivery is furthered by the number of potential delivery methods, the different forms in which CRISPR can be delivered as a therapeutic, and the disease target and tissue type in question. There are major challenges and limitations to delivery strategies, and it is imperative that future directions are guided by well-conducted studies that consider the full effect these variables have on the eventual outcome. In this review we will discuss the advances of the latest in vivo CRISPR/Cas9 delivery strategies and highlight the challenges yet to be overcome.


Assuntos
Edição de Genes , Terapia Genética/métodos , Sistemas CRISPR-Cas , Humanos
9.
Chem Soc Rev ; 47(10): 3421-3432, 2018 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-29537040

RESUMO

The delivery of proteins into cells is a potential game changer for a wide array of therapeutic purposes, including cancer therapy, immunomodulation and treatment of inherited diseases. In this review, we present recently developed nanoassemblies for protein delivery that utilize strategies that range from direct assembly, encapsulation and composite formation. We will discuss factors that affect the efficacy of nanoassemblies for delivery from the perspective of both nanoparticles and proteins. Challenges in the field, particularly achieving effective cytosolar protein delivery through endosomal escape or evasion are discussed.


Assuntos
Nanopartículas/metabolismo , Proteínas/metabolismo , Linhagem Celular , Humanos , Substâncias Macromoleculares/síntese química , Substâncias Macromoleculares/química , Substâncias Macromoleculares/metabolismo , Nanopartículas/química , Proteínas/química
11.
Nanoscale ; 15(24): 10351-10359, 2023 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-37288531

RESUMO

Vaccination through cellular transfection of nucleotide-based vaccines is a powerful approach to combatting disease. Plasmid DNA (pDNA) vaccines are particularly promising vectors for non-viral immunomodulation that afford high degrees of potency and flexibility. Versatile guanidinium-functionalized poly(oxanorbornene)imide (PONI-Guan) homopolymers were used to facilitate non-disruptive pDNA condensation into discrete polyplexes, enabling efficient in vitro transfection of endothelial cells and HD-11 macrophages. Translation of these vectors for vaccination of white leghorn chickens against Newcastle disease virus (NDV) elicited strong humoral immune responses against the virus. This approach presents a highly versatile method for targeted immunomodulation in vivo, with the potential for translatability as a non-viral vaccine platform.


Assuntos
Galinhas , Polímeros , Animais , Galinhas/genética , Células Endoteliais , Plasmídeos/genética , DNA/genética , Vacinação
12.
Biomaterials ; 302: 122344, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37857021

RESUMO

Intracellular pathogenic bacteria use immune cells as hosts for bacterial replication and reinfection, leading to challenging systemic infections including peritonitis. The spread of multidrug-resistant (MDR) bacteria and the added barrier presented by host cell internalization limit the efficacy of standard antibiotic therapies for treating intracellular infections. We present a non-antibiotic strategy to treat intracellular infections. Antimicrobial phytochemicals were stabilized and delivered by polymer-stabilized biodegradable nanoemulsions (BNEs). BNEs were fabricated using different phytochemicals, with eugenol-loaded BNEs (E-BNEs) affording the best combination of antimicrobial efficacy, macrophage accumulation, and biocompatibility. The positively-charged polymer groups of the E-BNEs bind to the cell surface of macrophages, facilitating the entry of eugenol that then kills the intracellular bacteria without harming the host cells. Confocal imaging and flow cytometry confirmed that this entry occurred mainly via cholesterol-dependent membrane fusion. As eugenol co-localized and interacted with intracellular bacteria, antibacterial efficacy was maintained. E-BNEs reversed the immunosuppressive effects of MRSA on macrophages. Notably, E-BNEs did not elicit resistance selection after multiple exposures of MRSA to sub-therapeutic doses. The E-BNEs were highly effective against a murine model of MRSA-induced peritonitis with better bacterial clearance (99 % bacteria reduction) compared to clinically-employed treatment with vancomycin. Overall, these findings demonstrate the potential of E-BNEs in treating peritonitis and other refractory intracellular infections.


Assuntos
Anti-Infecciosos , Staphylococcus aureus Resistente à Meticilina , Peritonite , Camundongos , Animais , Eugenol/farmacologia , Eugenol/uso terapêutico , Anti-Infecciosos/farmacologia , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Polímeros/farmacologia , Peritonite/tratamento farmacológico , Peritonite/microbiologia , Testes de Sensibilidade Microbiana
13.
ACS Nano ; 17(5): 4315-4326, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36802503

RESUMO

Uncontrolled inflammation is responsible for acute and chronic diseases in the lung. Regulating expression of pro-inflammatory genes in pulmonary tissue using small interfering RNA (siRNA) is a promising approach to combatting respiratory diseases. However, siRNA therapeutics are generally hindered at the cellular level by endosomal entrapment of delivered cargo and at the organismal level by inefficient localization in pulmonary tissue. Here we report efficient anti-inflammatory activity in vitro and in vivo using polyplexes of siRNA and an engineered cationic polymer (PONI-Guan). PONI-Guan/siRNA polyplexes efficiently deliver siRNA cargo to the cytosol for highly efficient gene knockdown. Significantly, these polyplexes exhibit inherent targeting to inflamed lung tissue following intravenous administration in vivo. This strategy achieved effective (>70%) knockdown of gene expression in vitro and efficient (>80%) silencing of TNF-α expression in lipopolysaccharide (LPS)-challenged mice using a low (0.28 mg/kg) siRNA dosage.


Assuntos
Pneumonia , Polímeros , Animais , Camundongos , RNA Interferente Pequeno , Polímeros/metabolismo , RNA de Cadeia Dupla/metabolismo , Endossomos/metabolismo , Pneumonia/terapia , Pneumonia/metabolismo
14.
Chem Sci ; 13(43): 12899-12905, 2022 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-36519060

RESUMO

Cell surface glycans serve fundamental roles in many biological processes, including cell-cell interaction, pathogen infection, and cancer metastasis. Cancer cell surface have alternative glycosylation to healthy cells, making these changes useful hallmarks of cancer. However, the diversity of glycan structures makes glycosylation profiling very challenging, with glycan 'fingerprints' providing an important tool for assessing cell state. In this work, we utilized the pH-responsive differential binding of boronic acid (BA) moieties with cell surface glycans to generate a high-content six-channel BA-based sensor array that uses a single polymer to distinguish mammalian cell types. This sensing platform provided efficient discrimination of cancer cells and readily discriminated between Chinese hamster ovary (CHO) glycomutants, providing evidence that discrimination is glycan-driven. The BA-functionalized polymer sensor array is readily scalable, providing access to new diagnostic and therapeutic strategies for cell surface glycosylation-associated diseases.

15.
Chem Sci ; 13(41): 12071-12077, 2022 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-36349111

RESUMO

Bioorthogonal catalysis mediated by transition metal catalysts (TMCs) presents a versatile tool for in situ generation of diagnostic and therapeutic agents. The use of 'naked' TMCs in complex media faces numerous obstacles arising from catalyst deactivation and poor water solubility. The integration of TMCs into engineered inorganic scaffolds provides 'nanozymes' with enhanced water solubility and stability, offering potential applications in biomedicine. However, the clinical translation of nanozymes remains challenging due to their side effects including the genotoxicity of heavy metal catalysts and unwanted tissue accumulation of the non-biodegradable nanomaterials used as scaffolds. We report here the creation of an all-natural catalytic "polyzyme", comprised of gelatin-eugenol nanoemulsion engineered to encapsulate catalytically active hemin, a non-toxic iron porphyrin. These polyzymes penetrate biofilms and eradicate mature bacterial biofilms through bioorthogonal activation of a pro-antibiotic, providing a highly biocompatible platform for antimicrobial therapeutics.

16.
JACS Au ; 2(7): 1679-1685, 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35911454

RESUMO

Macrophages migrate to tumor sites by following chemoattractant gradients secreted by tumor cells, providing a truly active targeting strategy for cancer therapy. However, macrophage-based delivery faces challenges of cargo loading, control of release, and effects of the payload on the macrophage vehicle. We present a strategy that employs bioorthogonal "nanozymes" featuring transition metal catalysts (TMCs) to provide intracellular "factories" for the conversion of prodyes and prodrugs into imaging agents and chemotherapeutics. These nanozymes solubilize and stabilize the TMCs by embedding them into self-assembled monolayer coating gold nanoparticles. Nanozymes delivered into macrophages were intracellularly localized and retained activity even after prolonged (72 h) incubation. Significantly, nanozyme-loaded macrophages maintained their inherent migratory ability toward tumor cell chemoattractants, efficiently killing cancer cells in cocultures. This work establishes the potential of nanozyme-loaded macrophages for tumor site activation of prodrugs, providing readily tunable dosages and delivery rates while minimizing off-target toxicity of chemotherapeutics.

17.
ACS Nano ; 16(5): 7323-7330, 2022 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-35435664

RESUMO

Current strategies for the delivery of proteins into cells face general challenges of endosomal entrapment and concomitant degradation of protein cargo. Efficient delivery directly to the cytosol overcomes this obstacle: we report here the use of biotin-streptavidin tethering to provide a modular approach to the generation of nanovectors capable of a cytosolic delivery of biotinylated proteins. This strategy uses streptavidin to organize biotinylated protein and biotinylated oligo(glutamate) peptide into modular complexes that are then electrostatically self-assembled with a cationic guanidinium-functionalized polymer. The resulting polymer-protein nanocomposites demonstrate efficient cytosolic delivery of six biotinylated protein cargos of varying size, charge, and quaternary structure. Retention of protein function was established through efficient cell killing via delivery of the chemotherapeutic enzyme granzyme A. This platform represents a versatile and modular approach to intracellular delivery through the noncovalent tethering of multiple components into a single delivery vector.


Assuntos
Biotina , Nanocompostos , Estreptavidina/química , Biotina/química , Citosol/metabolismo , Proteínas/química , Polímeros/química
18.
Nanoscale ; 14(6): 2411-2418, 2022 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-35089292

RESUMO

Control over supramolecular recognition between proteins and nanoparticles (NPs) is of fundamental importance in therapeutic applications and sensor development. Most NP-protein binding approaches use 'tags' such as biotin or His-tags to provide high affinity; protein surface recognition provides a versatile alternative strategy. Generating high affinity NP-protein interactions is challenging however, due to dielectric screening at physiological ionic strengths. We report here the co-engineering of nanoparticles and protein to provide high affinity binding. In this strategy, 'supercharged' proteins provide enhanced interfacial electrostatic interactions with complementarily charged nanoparticles, generating high affinity complexes. Significantly, the co-engineered protein-nanoparticle assemblies feature high binding affinity even at physiologically relevant ionic strength conditions. Computational studies identify both hydrophobic and electrostatic interactions as drivers for these high affinity NP-protein complexes.


Assuntos
Nanopartículas , Interações Hidrofóbicas e Hidrofílicas , Ligação Proteica , Proteínas , Eletricidade Estática
19.
Nat Nanotechnol ; 17(1): 86-97, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34795440

RESUMO

This study shows that the supramolecular arrangement of proteins in nanoparticle structures predicts nanoparticle accumulation in neutrophils in acute lung inflammation (ALI). We observed homing to inflamed lungs for a variety of nanoparticles with agglutinated protein (NAPs), defined by arrangement of protein in or on the nanoparticles via hydrophobic interactions, crosslinking and electrostatic interactions. Nanoparticles with symmetric protein arrangement (for example, viral capsids) had no selectivity for inflamed lungs. Flow cytometry and immunohistochemistry showed NAPs have tropism for pulmonary neutrophils. Protein-conjugated liposomes were engineered to recapitulate NAP tropism for pulmonary neutrophils. NAP uptake in neutrophils was shown to depend on complement opsonization. We demonstrate diagnostic imaging of ALI with NAPs; show NAP tropism for inflamed human donor lungs; and show that NAPs can remediate pulmonary oedema in ALI. This work demonstrates that structure-dependent tropism for neutrophils drives NAPs to inflamed lungs and shows NAPs can detect and treat ALI.


Assuntos
Inflamação/patologia , Pulmão/patologia , Nanopartículas/química , Neutrófilos/patologia , Proteínas/química , Doença Aguda , Aglutinação/efeitos dos fármacos , Animais , Anticorpos/farmacologia , Reagentes de Ligações Cruzadas/química , Dextranos/química , Humanos , Lipopolissacarídeos , Lipossomos , Pulmão/diagnóstico por imagem , Masculino , Camundongos Endogâmicos C57BL , Muramidase/metabolismo , Neutrófilos/efeitos dos fármacos , Proteínas Opsonizantes/metabolismo , Eletricidade Estática , Distribuição Tecidual/efeitos dos fármacos , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
20.
ACS Appl Mater Interfaces ; 13(40): 48301-48307, 2021 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-34606711

RESUMO

Nosocomial infections, caused by bacterial contamination of medical devices and implants, are a serious healthcare concern. We demonstrate here, the use of fluorous-cured protein nanofilm coatings for generating antimicrobial surfaces. In this approach, bacteria-repelling films are created by heat-curing proteins in fluorous media. These films are then loaded with antibiotics, with release controlled via electrostatic interactions between therapeutic and protein film building blocks to provide bactericidal surfaces. This film fabrication process is additive-free, biocompatible, biodegradable, and can be used to provide antimicrobial coatings for both three-dimensional (2D) and 3D objects for use in indwelling devices.


Assuntos
Antibacterianos/farmacologia , Incrustação Biológica/prevenção & controle , Materiais Revestidos Biocompatíveis/química , Preparações de Ação Retardada/química , Animais , Antibacterianos/química , Bovinos , Colistina/química , Colistina/farmacologia , Liberação Controlada de Fármacos , Fluoresceína/química , Corantes Fluorescentes/química , Fluorocarbonos/química , Próteses e Implantes , Pseudomonas aeruginosa/efeitos dos fármacos , Rodamina 123/química , Soroalbumina Bovina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA