Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Adv Mater ; 36(4): e2306704, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37947789

RESUMEN

Cells rely on secreted signaling molecules to coordinate essential biological functions including development, metabolism, and immunity. Unfortunately, such signaling processes remain difficult to measure with sufficient chemical specificity and temporal resolution. To address this need, an aptamer-conjugated hydrogel matrix that enables continuous fluorescent measurement of specific secreted analytes - in two dimensions, in real-time is developed. As a proof of concept, real-time imaging of inter-cellular cyclic adenosine 3',5'-monophosphate (cAMP) signals in Dictyostelium discoideum amoeba cells is performed. A set of aptamer switches that generate a rapid and reversible change in fluorescence in response to cAMP signals is engineered. By combining multiple switches with different dynamic ranges, measure cAMP concentrations spanning three orders of magnitude in a single experiment can be measured. These sensors are embedded within a biocompatible hydrogel on which cells are cultured and their cAMP secretions can be imaged using fluorescent microscopy. Using this aptamer-hydrogel material system, the first direct measurements of oscillatory cAMP signaling that correlate closely with previous indirect measurements are achieved. Using different aptamer switches, this approach can be generalized for measuring other secreted molecules to directly visualize diverse extracellular signaling processes and the biological effects that they trigger in recipient cells.


Asunto(s)
AMP Cíclico , Dictyostelium , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Dictyostelium/metabolismo , Hidrogeles/metabolismo , Transducción de Señal , Adenosina/metabolismo , Oligonucleótidos
2.
Proc Natl Acad Sci U S A ; 120(52): e2313009120, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38109533

RESUMEN

Genetic medicines have the potential to treat various diseases; however, certain ailments including inflammatory diseases and cancer would benefit from control over extracellular localization of therapeutic proteins. A critical gap therefore remains the need to develop and incorporate methodologies that allow for posttranslational control over expression dynamics, localization, and stability of nucleic acid-generated protein therapeutics. To address this, we explored how the body's endogenous machinery controls protein localization through signal peptides (SPs), including how these motifs could be incorporated modularly into therapeutics. SPs serve as a virtual zip code for mRNA transcripts that direct the cell where to send completed proteins within the cell and the body. Utilizing this signaling biology, we incorporated secretory SP sequences upstream of mRNA transcripts coding for reporter, natural, and therapeutic proteins to induce secretion of the proteins into systemic circulation. SP sequences generated secretion of various engineered proteins into the bloodstream following intravenous, intramuscular, and subcutaneous SP mRNA delivery by lipid, polymer, and ionizable phospholipid delivery carriers. SP-engineered etanercept/TNF-α inhibitor proteins demonstrated therapeutic efficacy in an imiquimod-induced psoriasis model by reducing hyperkeratosis and inflammation. An SP-engineered anti-PD-L1 construct mediated mRNA encoded proteins with longer serum half-lives that reduced tumor burden and extended survival in MC38 and B16F10 cancer models. The modular nature of SP platform should enable intracellular and extracellular localization control of various functional proteins for diverse therapeutic applications.


Asunto(s)
Dermatitis , Melanoma , Psoriasis , Humanos , Animales , Melanoma/tratamiento farmacológico , Melanoma/genética , Psoriasis/tratamiento farmacológico , Psoriasis/genética , Inflamación/patología , Señales de Clasificación de Proteína , ARN Mensajero/genética , Modelos Animales de Enfermedad
3.
Nat Commun ; 14(1): 7322, 2023 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-37951948

RESUMEN

Approximately 10% of Cystic Fibrosis (CF) patients, particularly those with CF transmembrane conductance regulator (CFTR) gene nonsense mutations, lack effective treatments. The potential of gene correction therapy through delivery of the CRISPR/Cas system to CF-relevant organs/cells is hindered by the lack of efficient genome editor delivery carriers. Herein, we report improved Lung Selective Organ Targeting Lipid Nanoparticles (SORT LNPs) for efficient delivery of Cas9 mRNA, sgRNA, and donor ssDNA templates, enabling precise homology-directed repair-mediated gene correction in CF models. Optimized Lung SORT LNPs deliver mRNA to lung basal cells in Ai9 reporter mice. SORT LNP treatment successfully corrected the CFTR mutations in homozygous G542X mice and in patient-derived human bronchial epithelial cells with homozygous F508del mutations, leading to the restoration of CFTR protein expression and chloride transport function. This proof-of-concept study will contribute to accelerating the clinical development of mRNA LNPs for CF treatment through CRISPR/Cas gene correction.


Asunto(s)
Fibrosis Quística , Humanos , Ratones , Animales , Fibrosis Quística/terapia , Fibrosis Quística/tratamiento farmacológico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Sistemas CRISPR-Cas/genética , ARN Guía de Sistemas CRISPR-Cas , Pulmón/metabolismo , ARN Mensajero/genética , ARN Mensajero/uso terapéutico
4.
J Control Release ; 361: 361-372, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37536547

RESUMEN

Messenger RNA (mRNA) can treat genetic disease using protein replacement or genome editing approaches but requires a suitable carrier to circumnavigate biological barriers and access the desired cell type within the target organ. Lipid nanoparticles (LNPs) are widely used in the clinic for mRNA delivery yet are limited in their applications due to significant hepatic accumulation because of the formation of a protein corona enriched in apolipoprotein E (ApoE). Our lab developed selective organ targeting (SORT) LNPs that incorporate a supplementary component, termed a SORT molecule, for tissue-specific mRNA delivery to the liver, spleen, and lungs of mice. Mechanistic work revealed that the biophysical class of SORT molecule added to the LNP forms a distinct protein corona that helps determine where in the body mRNA is delivered. To better understand which plasma proteins could drive tissue-specific mRNA delivery, we characterized a panel of quaternary ammonium lipids as SORT molecules to assess how chemical structure affects the organ-targeting outcomes and protein corona of lung-targeting SORT LNPs. We discovered that variations in the chemical structure of both the lipid alkyl tail and headgroup impact the potency and specificity of mRNA delivery to the lungs. Furthermore, changes to the chemical structure alter the quantities and identities of protein corona constituents in a manner that correlates with organ-targeting outcomes, with certain proteins appearing to promote lung targeting whereas others reduce delivery to off-target organs. These findings unveil a nuanced relationship between LNP chemistry and endogenous targeting, where the ensemble of proteins associated with an LNP can play various roles in determining the tissue-specificity of mRNA delivery, providing further design criteria for optimization of clinically-relevant nanoparticles for extrahepatic delivery of genetic payloads.


Asunto(s)
Compuestos de Amonio , Nanopartículas , Corona de Proteínas , Ratones , Animales , Lípidos/química , ARN Mensajero/metabolismo , Liposomas , Nanopartículas/química , ARN Interferente Pequeño/química
5.
Nat Protoc ; 18(1): 265-291, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36316378

RESUMEN

A new methodology termed selective organ targeting (SORT) was recently developed that enables controllable delivery of nucleic acids to target tissues. SORT lipid nanoparticles (LNPs) involve the inclusion of SORT molecules that accurately tune delivery to the liver, lungs and spleen of mice after intravenous administration. Nanoparticles can be engineered to target specific cells and organs in the body by passive, active and endogenous targeting mechanisms that require distinct design criteria. SORT LNPs are modular and can be prepared using scalable, synthetic chemistry and established engineering formulation methods. This protocol provides detailed procedures, including the synthesis of a representative ionizable cationic lipid, preparation of multiple classes of SORT LNPs by pipette, vortex and microfluidic mixing methods, physical characterization, and in vitro/in vivo mRNA delivery evaluation. Depending on the scale of the experiments, the synthesis of the ionizable lipid requires 4-6 d; LNPs can be formulated within several hours; LNP characterization can be completed in 2-4 h; and in vitro/in vivo evaluation studies require 1-14 d, depending on the design and application. Our strategy offers a versatile and practical method for rationally designing nanoparticles that accurately target specific organs. The SORT LNPs generated as described in this protocol can therefore be applied to multiple classes of LNP systems for therapeutic nucleic acid delivery and facilitate the development of protein replacement and genetic medicines in target tissues. This protocol does not require specific expertise, is modular to various lipids within defined physicochemical classes, and should be accomplishable by researchers from various backgrounds.


Asunto(s)
Liposomas , Nanopartículas , Ratones , Animales , ARN Mensajero/química , Nanopartículas/química , Lípidos/química , ARN Interferente Pequeño/genética
6.
Small ; 14(47): e1802709, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30222252

RESUMEN

Understanding the key factors for successful subcellular compartment targeting for cargo delivery systems is of great interest in a variety of fields such as bionanotechnology, cell biology, and nanotherapies. However, the fundamental basis for intracellular transportation with these systems has thus far rarely been discussed. As a cargo vector, porous coordination cages (PCCs) have great potential for use in cancer nanotherapy and to elucidate fundamental insight regarding subcellular compartment targeting. Herein, it is shown that the transportation of PCC cargo vectors though various subcellular barriers of the mammalian cell can be manipulated by tuning the vector's electronic property and surface affinity. It is found that the PCCs become selectively aggregated at the cell membrane, the cytoplasm, or the nucleus, respectively. When a DNA topoisomerase inhibitor is delivered into the nucleus by a neutral and lipophilic PCC, the anticancer efficacy is dramatically improved. The findings shed light to tune the interactions at the "bio-nano" interface. This study provides a key strategy for future work in targeting specific cell organelles for cell imaging, cargo delivery, and therapy. This research also offers key insight into the engineering of nanoscopic materials for furnishing cell organelle-specificity.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Animales , Biotecnología/métodos , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Porosidad , Inhibidores de Topoisomerasa
7.
Angew Chem Int Ed Engl ; 57(20): 5725-5730, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29536600

RESUMEN

Prodrug activation, by exogenously administered enzymes, for cancer therapy is an approach to achieve better selectivity and less systemic toxicity than conventional chemotherapy. However, the short half-lives of the activating enzymes in the bloodstream has limited its success. Demonstrated here is that a tyrosinase-MOF nanoreactor activates the prodrug paracetamol in cancer cells in a long-lasting manner. By generating reactive oxygen species (ROS) and depleting glutathione (GSH), the product of the enzymatic conversion of paracetamol is toxic to drug-resistant cancer cells. Tyrosinase-MOF nanoreactors cause significant cell death in the presence of paracetamol for up to three days after being internalized by cells, while free enzymes totally lose activity in a few hours. Thus, enzyme-MOF nanocomposites are envisioned to be novel persistent platforms for various biomedical applications.


Asunto(s)
Acetaminofén/farmacología , Antineoplásicos/farmacología , Estructuras Metalorgánicas/metabolismo , Monofenol Monooxigenasa/metabolismo , Nanopartículas/metabolismo , Acetaminofén/química , Acetaminofén/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Estructuras Metalorgánicas/química , Modelos Moleculares , Estructura Molecular , Monofenol Monooxigenasa/química , Nanopartículas/química , Tamaño de la Partícula , Relación Estructura-Actividad , Propiedades de Superficie
8.
Angew Chem Int Ed Engl ; 57(19): 5283-5287, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29431893

RESUMEN

Earth-abundant first-row transition-metal nanoclusters (NCs) have been extensively investigated as catalysts. However, their catalytic activity is relatively low compared with noble metal NCs. Enhanced catalytic activity of cobalt NCs can be achieved by encapsulating Co NCs in soluble porous coordination cages (PCCs). Two cages, PCC-2a and 2b, possess almost identical cavity in shape and size, while PCC-2a has five times more net charges than PCC-2b. Co2+ cations were accumulated in PCC-2a and reduced to ultra-small Co NCs in situ, while for PCC-2b, only bulky Co particles were formed. As a result, Co NCs@PCC-2a accomplished the highest catalytic activity in the hydrolysis of ammonium borane among all the first-row transition-metals NCs. Based on these results, it is envisioned that confining in the charged porous coordination cage could be a novel route for the synthesis of ultra-small NCs with extraordinary properties.

9.
Nat Commun ; 8(1): 2075, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29234027

RESUMEN

Enhancing or restoring enzymatic function in cells is highly desirable in applications ranging from ex vivo cellular manipulations to enzyme replacement therapies in humans. However, because enzymes degrade in biological milieus, achieving long-term enzymatic activities can be challenging. Herein we report on the in cellulo properties of nanofactories that consist of antioxidative enzymes encapsulated in metal-organic frameworks (MOFs). We demonstrate that, while free enzymes display weak activities for only a short duration, these efficient nanofactories protect human cells from toxic reactive oxygen species for up to a week. Remarkably, these results are obtained in spite of the nanofactories being localized in lysosomes, acidic organelles that contain a variety of proteases. The long-term persistence of the nanofactories is attributed to the chemical stability of MOF in low pH environment and to the protease resistance provided by the protective cage formed by the MOF around the encapsulated enzymes.


Asunto(s)
Citoplasma/metabolismo , Endopeptidasas/metabolismo , Estructuras Metalorgánicas/metabolismo , Nanotecnología/métodos , Orgánulos/metabolismo , Citoplasma/efectos de los fármacos , Endopeptidasas/química , Pruebas de Enzimas , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Estructuras Metalorgánicas/química , Orgánulos/efectos de los fármacos , Estrés Oxidativo , Especies Reactivas de Oxígeno/toxicidad
10.
Chem Soc Rev ; 46(11): 3386-3401, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28451673

RESUMEN

The ex vivo application of enzymes in various processes, especially via enzyme immobilization techniques, has been extensively studied in recent years in order to enhance the recyclability of enzymes, to minimize enzyme contamination in the product, and to explore novel horizons for enzymes in biomedical applications. Possessing remarkable amenability in structural design of the frameworks as well as almost unparalelled surface tunability, Metal-Organic Frameworks (MOFs) have been gaining popularity as candidates for enzyme immobilization platforms. Many MOF-enzyme composites have achieved unprecedented results, far outperforming free enzymes in many aspects. This review summarizes recent developments of MOF-enzyme composites with special emphasis on preparative techniques and the synergistic effects of enzymes and MOFs. The applications of MOF-enzyme composites, primarily in transferation, catalysis and sensing, are presented as well. The enhancement of enzymatic activity of the composites over free enzymes in biologically incompatible conditions is emphasized in many cases.


Asunto(s)
Enzimas/química , Estructuras Metalorgánicas/química , Enzimas/metabolismo , Estructuras Metalorgánicas/metabolismo , Propiedades de Superficie
11.
J Am Chem Soc ; 138(16): 5299-307, 2016 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-27045822

RESUMEN

Quests for advanced functionalities in metal-organic frameworks (MOFs) inevitably encounter increasing complexity in their tailored framework architectures, accompanied by heightened challenges with their geometric design. In this paper, we demonstrate the feasibility of rationally exploiting topological prediction as a blueprint for predesigned MOFs. A new triangular frusta secondary building unit (SBU), {Zn4(tz)3}, was bridged by three TDC(2-) fragments to initially form a trigonal prismatic node, {Zn8(tz)6(TDC)3} (Htz = 1H-1,2,3-triazole and H2TDC = 2,5-thiophenedicarboxylic acid). Furthermore, the trigonal prism unit can be considered as a double SBU derived from triply bound triangular frusta. By considering theoretical derived nets for linking this trigonal-prismatic node with ditopic, tritopic, and tetratopic linkers, we have synthesized and characterized a new family of MOFs that adopt the decorated lon, jea, and xai nets, respectively. Pore sizes have also been successively increased within TPMOF-n family, which facilitates heterogeneous biomimetic catalysis with Fe-porphyrin-based TPMOF-7 as a catalyst.

12.
Chem Sci ; 7(12): 6969-6973, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28451131

RESUMEN

A hierarchical porous metal-organic framework (MOF), PCN-888, containing three types of cavities was utilized to couple two enzymes into a tandem nanoreactor. The largest cavity (6.2 nm) can only accommodate one molecule of glucose oxidase (GOx). The intermediate cavity (5.0 nm) can accommodate one and only one molecule of horseradish peroxidase (HRP). The small cavity (2.0 nm) has sufficient size for neither GOx nor HRP, and remains open as a substrate diffusion pathway. The coupling of the two enzymes can only be achieved through a unique stepwise encapsulation with a specific order (GOx first, followed by HRP). The tandem nanoreactor shows excellent catalytic performances and negligible enzyme leaching. Its catalytic activity is well maintained within several catalytic cycles. Moreover, PCN-888 can provide protection to the encapsulated enzymes against trypsin digestion, indicating the in vitro and in vivo stability of the nanoreactor.

13.
Angew Chem Int Ed Engl ; 54(49): 14696-700, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26494126

RESUMEN

Cooperative cluster metalation and ligand migration were performed on a Zr-MOF, leading to the isolation of unique bimetallic MOFs based on decanuclear Zr6M4 (M = Ni, Co) clusters. The M(2+) reacts with the µ3-OH and terminal H2O ligands on an 8-connected [Zr6O4(OH)8(H2O)4] cluster to form a bimetallic [Zr6M4O8(OH)8(H2O)8] cluster. Along with the metalation of Zr6 cluster, ligand migration is observed in which a Zr-carboxylate bond dissociates to form a M-carboxylate bond. Single-crystal to single-crystal transformation is realized so that snapshots for cooperative cluster metalation and ligand migration processes are captured by successive single-crystal X-ray structures. In(3+) was metalated into the same Zr-MOF which showed excellent catalytic activity in the acetaldehyde cyclotrimerization reaction. This work not only provides a powerful tool to functionalize Zr-MOFs with other metals, but also structurally elucidates the formation mechanism of the resulting heterometallic MOFs.

14.
Nat Commun ; 6: 5979, 2015 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-25598311

RESUMEN

Enzymatic catalytic processes possess great potential in chemical manufacturing, including pharmaceuticals, fuel production and food processing. However, the engineering of enzymes is severely hampered due to their low operational stability and difficulty of reuse. Here, we develop a series of stable metal-organic frameworks with rationally designed ultra-large mesoporous cages as single-molecule traps (SMTs) for enzyme encapsulation. With a high concentration of mesoporous cages as SMTs, PCN-333(Al) encapsulates three enzymes with record-high loadings and recyclability. Immobilized enzymes that most likely undergo single-enzyme encapsulation (SEE) show smaller Km than free enzymes while maintaining comparable catalytic efficiency. Under harsh conditions, the enzyme in SEE exhibits better performance than free enzyme, showing the effectiveness of SEE in preventing enzyme aggregation or denaturation. With extraordinarily large pore size and excellent chemical stability, PCN-333 may be of interest not only for enzyme encapsulation, but also for entrapment of other nanoscaled functional moieties.


Asunto(s)
Enzimas Inmovilizadas/química , Catálisis , Nanotecnología/métodos
15.
Chem Sci ; 6(12): 7044-7048, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29861943

RESUMEN

Kinetic labilization of the Fe(iii)-O coordination bond in a mesoporous metal-organic framework, PCN-333-Fe(iii), is realized by the reduction of Fe(iii) by Cr(ii). The Fe(ii)-Cr(ii) metathesis is thus completely achieved followed by this reductive labilization process that generates PCN-333-Cr(iii). The kinetic inertness of Cr(iii) provides PCN-333-Cr(iii) with enhanced chemical stability as well as a broader range of applications compared to those of PCN-333-Fe(iii). For instance, alkylamine incorporated PCN-333-Cr(iii) demonstrated significant carbon dioxide uptake while PCN-333-Fe(iii) lost its crystallinity after alkylamine treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA