Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(27): e2302367120, 2023 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-37364107

RESUMEN

Antimicrobial susceptibility testing plays a pivotal role in the discovery of new antibiotics. However, the development of simple, sensitive, and rapid assessment approaches remains challenging. Herein, we report an activated alkyne-based cascade signal amplification strategy for ultrafast and high-throughput antibiotic screening. First of all, a novel water-soluble aggregation-induced emission (AIE) luminogen is synthesized, which contains an activated alkyne group to enable fluorescence turn-on and metal-free click bioconjugation under physiological conditions. Taking advantage of the in-house established method for bacterial lysis, a number of clickable biological substances (i.e., bacterial solutes and debris) are released from the bacterial bodies, which remarkably increases the quantity of analytes. By means of the activated alkyne-mediated turn-on click bioconjugation, the system fluorescence signal is significantly amplified due to the increased labeling sites as well as the AIE effect. Such a cascade signal amplification strategy efficiently improves the detection sensitivity and thus enables ultrafast antimicrobial susceptibility assessment. By integration with a microplate reader, this approach is further applied to high-throughput antibiotic screening.


Asunto(s)
Alquinos , Antibacterianos , Antibacterianos/farmacología , Fluorescencia , Química Clic/métodos , Azidas
2.
Small ; 19(3): e2206657, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36394193

RESUMEN

Developing nature-inspired nanomaterials with enzymatic activity is essential in combating bacterial biofilms. Here, it is reported that incorporating the carboxylic acid in phenolic/Fe nano-networks can efficiently manipulate their peroxidase-like activity via the acidic microenvironment and neighboring effect of the carboxyl group. The optimal gallic acid/Fe (GA/Fe) nano-networks demonstrate highly enzymatic activity in catalyzing H2 O2 into oxidative radicals, damaging the cell membrane and extracellular DNA in Streptococcus mutans biofilms. Theoretical calculation suggests that the neighboring carboxyl group can aid the H2 O2 adsorption, free radical generation, and catalyst reactivation, resulting in superb catalytic efficiency. Further all-atom simulation suggests the peroxidation of lipids can increase the cell membrane fluidity and permeability. Also, GA/Fe nano-networks show great potential in inhibiting tooth decay and treating other biofilm-associated diseases without affecting the commensal oral flora. This strategy provides a facile and scale-up way to prepare the enzyme-like materials and manipulate their enzymatic activity for biomedical applications.


Asunto(s)
Peroxidasa , Streptococcus mutans , Peroxidasa/metabolismo , Streptococcus mutans/genética , Streptococcus mutans/metabolismo , Biopelículas
3.
Biomacromolecules ; 24(11): 5230-5244, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37733485

RESUMEN

As an acute ophthalmic infection, bacterial keratitis (BK) can lead to severe visual morbidity, such as corneal perforation, intraocular infection, and permanent corneal opacity, if rapid and effective treatments are not available. In addition to eradicating pathogenic bacteria, protecting corneal tissue from oxidative damage and promoting wound healing by relieving inflammation are equally critical for the efficient treatment of BK. Besides, it is very necessary to improve the bioavailability of drugs by enhancing the ocular surface adhesion and corneal permeability. In this investigation, therefore, a synergistic antibiotic-antioxidant treatment of BK was achieved based on multifunctional block copolymer vesicles, within which ciprofloxacin (CIP) was simultaneously encapsulated during the self-assembly. Due to the phenylboronic acid residues in the corona layer, these vesicles exhibited enhanced muco-adhesion, deep corneal epithelial penetration, and bacteria-targeting, which facilitated the drug delivery to corneal bacterial infection sites. Additionally, the abundant thioether moieties in the hydrophobic membrane enabled the vesicles to both have ROS-scavenging capacity and accelerated CIP release at the inflammatory corneal tissue. In vivo experiments on a mice model demonstrated that the multifunctional polymer vesicles achieved efficient treatment of BK, owing to the enhanced corneal adhesion and penetration, bacteria targeting, ROS-triggered CIP release, and the combined antioxidant-antibiotic therapy. This synergistic strategy holds great potential in the treatment of BK and other diseases associated with bacterial infections.


Asunto(s)
Infecciones Bacterianas del Ojo , Queratitis , Animales , Ratones , Antioxidantes/farmacología , Polímeros/química , Especies Reactivas de Oxígeno , Queratitis/tratamiento farmacológico , Queratitis/microbiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Ciprofloxacina , Infecciones Bacterianas del Ojo/tratamiento farmacológico , Infecciones Bacterianas del Ojo/microbiología
4.
J Am Chem Soc ; 144(21): 9312-9323, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35587998

RESUMEN

Self-sorting is a common phenomenon in eukaryotic cells and represents one of the versatile strategies for the formation of advanced functional materials; however, developing artificial self-sorting assemblies within living cells remains challenging. Here, we report on the GSH-responsive in situ self-sorting peptide assemblies within cancer cells for simultaneous organelle targeting to promote combinatorial organelle dysfunction and thereby cell death. The self-sorting system was created via the design of two peptides E3C16E6 and EVMSeO derived from lipid-inspired peptide interdigitating amphiphiles and peptide bola-amphiphiles, respectively. The distinct organization patterns of the two peptides facilitate their GSH-induced self-sorting into isolated nanofibrils as a result of cleavage of disulfide-connected hydrophilic domains or reduction of selenoxide groups. The GSH-responsive in situ self-sorting in the peptide assemblies within HeLa cells was directly characterized by super-resolution structured illumination microscopy. Incorporation of the thiol and ER-targeting groups into the self-sorted assemblies endows their simultaneous targeting of endoplasmic reticulum and Golgi apparatus, thus leading to combinatorial organelle dysfunction and cell death. Our results demonstrate the establishment of the in situ self-sorting peptide assemblies within living cells, thus providing a unique platform for drug targeting delivery and an alternative strategy for modulating biological processes in the future.


Asunto(s)
Aparato de Golgi , Péptidos , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Péptidos/química , Transporte de Proteínas
5.
J Am Chem Soc ; 144(15): 6907-6917, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35388694

RESUMEN

Enzyme-regulated in situ self-assembly of peptides represents one versatile strategy in the creation of theranostic agents, which, however, is limited by the strong dependence on enzyme overexpression. Herein, we reported the self-amplifying assembly of peptides precisely in macrophages associated with enzyme expression for improving the anti-inflammatory efficacy of conventional drugs. The self-amplifying assembling system was created via coassembling an enzyme-responsive peptide with its derivative functionalized with a protein ligand. Reduction of the peptides by the enzyme NAD(P)H quinone dehydrogenase 1 (NQO1) led to the formation of nanofibers with high affinity to the protein, thereby facilitating NQO1 expression. The improved NQO1 level conversely promoted the assembly of the peptides into nanofibers, thus establishing an amplifying relationship between the peptide assembly and the NQO1 expression in macrophages. Utilization of the amplifying assembling system as vehicles for drug dexamethasone allowed for its passive targeting delivery to acute injured lungs. Both in vitro and in vivo studies confirmed the capability of the self-amplifying assembling system to enhance the anti-inflammatory efficacy of dexamethasone via simultaneous alleviation of the reactive oxygen species side effect and downregulation of proinflammatory cytokines. Our findings demonstrate the manipulation of the assembly of peptides in living cells with a regular enzyme level via a self-amplification process, thus providing a unique strategy for the creation of supramolecular theranostic agents in living cells.


Asunto(s)
Nanofibras , Péptidos , Dexametasona , Ligandos , Macrófagos/metabolismo , Nanofibras/química , Péptidos/química
6.
Small ; 18(32): e2203100, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35843873

RESUMEN

Personalized cancer vaccination using nanomaterials holds great potential for cancer immunotherapy. Here, a nanochaperone (PBA-nChap) is tailored for in situ capture of tumor-associated antigens (TAAs) to improve cancer immunotherapy. The PBA-nChap is capable of i) efficiently capturing TAAs in situ; ii) protecting TAAs from degradation; iii) transporting TAAs to antigen-presenting cells and promoting cross-presentation. Intratumor injection of PBA-nChap in combination with pretreatment with photodynamic therapy (PDT) significantly enhances immune response and exhibits excellent antitumor efficacy. Moreover, nanovaccine prepared by simply co-culturing PBA-nChap with tumor cell fragments from surgery resected primary tumor in vitro synergized with immune checkpoint blockade (ICB) therapy can effectively inhibit tumor recurrence and metastasis after an operation. This work provides a promising platform for personalized cancer vaccination.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Fotoquimioterapia , Animales , Antígenos de Neoplasias , Humanos , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia
7.
Nano Lett ; 21(13): 5730-5737, 2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-34142834

RESUMEN

Mitochondrion-targeting therapy exhibits great potential in cancer therapy but significantly suffers from limited therapeutic efficiency. Here we report on mitochondrion-targeting supramolecular antagonist-inducing tumor cell death via simultaneously promoting cellular apoptosis and preventing survival. The supramolecular antagonist was created via coassembly of a mitochondrion-targeting pentapeptide with its two derivatives functionalized with a BH3 domain or the drug camptothecin (CPT). While drug CPT released from the antagonist induced cellular apoptosis via decreasing the mitochondrial membrane potential, the BH3 domain prevented cellular survival through facilitating the association between the supramolecular antagonists and antiapoptotic proteins, thereby initiating mitochondrial permeabilization. Both in vitro and in vivo studies confirmed the combinatorial therapeutic effect arising from the BH3 domain and CPT drug within the supramolecular antagonist on cell death and thereby inhibiting tumor growth. Our findings demonstrate an efficient combinatorial mechanism for mitochondrial dysfunction, thus potentially serving as novel organelle-targeting medicines.


Asunto(s)
Apoptosis , Camptotecina , Camptotecina/farmacología , Mitocondrias
8.
Angew Chem Int Ed Engl ; 61(19): e202200192, 2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35229425

RESUMEN

Protein misassembly leads to the formation of dysfunctional and toxic molecular species relating to neurodegeneration in Parkinson's disease and Alzheimer's disease. Here, we tailored a nanochaperone (αS-nChap) for α-synuclein to regulate its assembly. The αS-nChap is capable of i) specifically recognizing α-synuclein; ii) dynamically capturing and stabilizing monomeric α-synuclein and retarding oligomerization; iii) tightly capturing oligomeric α-synuclein to prevent fibrillization; and iv) transporting α-synuclein oligomers to the lysosomal degradation system. The regulation of α-synuclein assembly by αS-nChap was studied in vitro. Moreover, the role of αS-nChap preventing α-synuclein pathology in cells and protecting neurons from apoptosis was investigated. The strategy of tailoring a nanochaperone to regulate aberrant assembly of pathogenic proteins provides important insights into protein misfolding diseases. We foresee that αS-nChap has therapeutic value for Parkinson's disease.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Parkinson , Enfermedad de Alzheimer/metabolismo , Humanos , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo
9.
J Am Chem Soc ; 143(34): 13854-13864, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34410694

RESUMEN

Design of endogenous stimuli-responsive amino acids allows for precisely modulating proteins or peptides under a biological microenvironment and thereby regulating their performance. Herein we report a noncanonical amino acid 2-nitroimidazol-1-yl alanine and explore its functions in creation of the nitroreductase (NTR)-responsive peptide-based supramolecular probes for efficient hypoxia imaging. On the basis of the reduction potential of the nitroimidazole unit, the amino acid was synthesized via the Mitsunobu reaction between 2-nitroimidazole and a serine derivate. We elucidated the relationship between the NTR-responsiveness of the amino acid and the structural feature of peptides involving a series of peptides. This eventually facilitates development of aromatic peptides undergoing NTR-responsive self-assembly by rationally optimizing the sequences. Due to the intrinsic role of 2-nitroimidazole in the fluorescence quench, we created a morphology-transformable supramolecular probe for imaging hypoxic tumor cells based on NTR reduction. We found that the resulting supramolecular probes penetrated into solid tumors, thus allowing for efficient fluorescence imaging of tumor cells in hypoxic regions. Our findings demonstrate development of a readily synthesized and versatile amino acid with exemplified properties in creating fluorescent peptide nanostructures responsive to a biological microenvironment, thus providing a powerful toolkit for synthetic biology and development of novel biomaterials.


Asunto(s)
Aminoácidos/metabolismo , Péptidos/metabolismo , Alanina/química , Alanina/metabolismo , Aminoácidos/química , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Colorantes Fluorescentes/química , Ratones , Microscopía Confocal , Nitroimidazoles/química , Nitrorreductasas/metabolismo , Imagen Óptica , Péptidos/química , Trasplante Homólogo
10.
Small ; 17(31): e2100578, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34190401

RESUMEN

Drug delivery systems (DDS) are extensively studied to improve the solubility, stability, pharmacokinetic, and biodistribution of chemotherapeutics. However, the drug delivery efficiency of traditional DDS is often limited by the complicated biological barriers in vivo. Herein, a multistage adaptive nanoparticle (MAN) that simultaneously overcomes multiple biological barriers to achieve tumor-targeted drug delivery with high efficiency is presented. MAN has a core-shell structure, in which both the core and the shell are made of responsive polymers. This structure allows MAN to present different surface properties to adapt to its surrounding biological microenvironment, thereby achieving enhanced stability in blood circulation, improved tumor accumulation and cellular internalization in tumor tissues, and effective release of drug in cells. With these unique characteristics, the MAN loaded with docetaxel achieves effective tumor suppression with reduced systemic toxicity. Furthermore, MAN can load almost any hydrophobic drugs, providing a general strategy for the tumor-targeted delivery of hydrophobic drugs to overcome the multiple biological barriers and improve the efficacy of chemotherapy.


Asunto(s)
Antineoplásicos , Nanopartículas , Antineoplásicos/farmacología , Línea Celular Tumoral , Docetaxel/farmacología , Sistemas de Liberación de Medicamentos , Humanos , Distribución Tisular
11.
Small ; 17(8): e2006223, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33522123

RESUMEN

Combination therapy based on molecular drugs and therapeutic genes provides an effective strategy for malignant tumor treatment. However, effective gene and drug combinations for cancer treatment are limited by the widespread antagonism between therapeutic genes and molecular drugs. Herein, a calixarene-embedded nanoparticle (CENP) is developed to co-deliver molecular drugs and therapeutic genes without compromising their biological functions, thereby achieving interference-free gene-drug combination cancer therapy. CENP is composed of a cationic polyplex core and an acid-responsive polymer shell, allowing CENP loading and delivering therapeutic genes with improved circulation stability and enhanced tumor accumulation. Moreover, the introduction of carboxylated azocalix[4]arene, which is a hypoxia-responsive calixarene derivatives, in the polyplex core endows CENP with the capability to load molecular drugs through the host-guest complexation as well as inhibit the interference between the drugs and genes by encapsulating the drugs into its cavity. By loading doxorubicin and a plasmid DNA-based CRISPR interference system that targets miR-21, CENP exhibits the significantly enhanced anti-tumor effects in mice. Considering the wide variety of calixarene derivatives, CENP can be adapted to deliver almost any combination of drugs and genes, providing the potential as a universal platform for the development of interference-free gene-drug combination cancer therapy.


Asunto(s)
Calixarenos , Nanopartículas , Neoplasias , Animales , Línea Celular Tumoral , Terapia Combinada , Doxorrubicina , Ratones
12.
Nano Lett ; 20(6): 4454-4463, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32401534

RESUMEN

Inspired by heat shock proteins (HSPs), a self-assembly nanochaperone (nChap) is developed as a novel nanovaccine for boosting antitumor immune responses. Taking advantage of HSP-like microdomains and surface-decorated mannose, this nChap can efficiently capture antigens and ferry them into the dendritic cells (DCs). Subsequently, the nChap can blast lysosomes by transforming the structure and property of surface microdomains, thereby promoting antigen escape and enhancing their cross-presentation in cytoplasm. As a result, the nChap-based nanovaccine can elicit both CD4+ and CD8+ T cell-based immune responses and shows an excellent preventive effect on melanoma. Further combination of the nanovaccine with antiprogrammed death-1 (anti-PD-1) checkpoint blockade offers effective inhibition on the growth of already-established melanoma. Therefore, this nC ap-based nanovaccine provides a simple and robust strategy in mimicking HSPs to realize structure-assisted antigen capture, surface-receptor-mediated DC internalization, and both activation of humoral immunity and cellular immunity, promising for efficient cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Proteínas de Choque Térmico , Inmunoterapia , Melanoma , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Humanos , Inmunidad , Melanoma/tratamiento farmacológico
13.
Nano Lett ; 20(3): 1755-1765, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32069419

RESUMEN

Insulin would undergo unfolding and fibrillation under stressed conditions, which may cause serious biotechnological and medical problems. Herein, by mimicking the structure and functions of natural chaperones HSP70s, self-assembled polymeric micelles are used as nanochaperones for the delivery of insulin. The confined hydrophobic domains on the surface of nanochaperones adsorb partially unfolded insulin, inhibiting the aggregation and fibrillation and enhancing the stability of insulin. The bioactivity of insulin is well-reserved after incubation with the nanochaperones at 37 °C for 7 d or heating at 70 °C for 1 h. The stealthy poly(ethylene glycol) chains around the confined domains protect the adsorbed insulin from enzymatic degradation and prolong the circulation time. More importantly, the excellent glucose sensitivity of the hydrophobic domains enables the nanochaperones to release and refold insulin in native form in response to hyperglycemia. This kind of nanochaperone may offer a hopeful strategy for the protection and delivery of insulin.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Insulina , Chaperonas Moleculares , Nanoestructuras , Animales , Diabetes Mellitus Experimental/metabolismo , Insulina/química , Insulina/farmacocinética , Insulina/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Chaperonas Moleculares/química , Chaperonas Moleculares/farmacocinética , Chaperonas Moleculares/farmacología , Células 3T3 NIH , Nanoestructuras/química , Nanoestructuras/uso terapéutico
14.
Angew Chem Int Ed Engl ; 60(19): 10865-10870, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33595165

RESUMEN

Nanochaperones have been designed and used for regulating the (re)folding of proteins, treating protein misfolding-related diseases, and, more recently, in drug delivery. Despite various successes, a complete understanding of the working mechanisms remains elusive, which represents a challenge for the realization of their full potential. Here, we thoroughly investigated the functioning of differently charged nanochaperones that regulate the refolding of thermally denatured lysozyme. We found that the balance between the capture and release of lysozyme clients that are controlled by nanochaperones plays a key role in regulating refolding. More importantly, the findings could be applied to other enzymes with various physicochemical properties. On the basis of these results, we could recover the activity of enzymes with high efficiency either after 20 days of storage at 40 °C or heating at high temperatures for 30-60 min. Our results provide important new design strategies for nanochaperone systems to improve their properties and expand their applications.


Asunto(s)
Chaperonas Moleculares/química , Muramidasa/química , Temperatura , Muramidasa/metabolismo , Tamaño de la Partícula , Desnaturalización Proteica , Pliegue de Proteína
15.
Angew Chem Int Ed Engl ; 60(32): 17714-17719, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34028150

RESUMEN

A lipid named DCPA was synthesized under microwave-assisted heating. DCPA possesses a pyridine betaine, hydrophilic group that can be complexed with water through hydrogen bonding (DCPA-H2 O). DCPA-H2 O liposomes became protonated relatively fast already at pH<6.8, due to the high HOMO binding energy of DCPA-H2 O. In murine models, DCPA-H2 O liposomes had longer blood circulation times than natural DPPC or cationic DCPM liposomes, while after tail-vein injection DCPA-H2 O liposomes targeted faster to solid tumors and intra-abdominal infectious biofilms. Therapeutic efficacy in a murine, infected wound-healing model of tail-vein injected ciprofloxacin-loaded DCPA-H2 O liposomes exceeded the ones of clinically applied ciprofloxacin as well as of ciprofloxacin-loaded DPPC or DCPM liposomes.


Asunto(s)
Portadores de Fármacos/farmacocinética , Liposomas/farmacocinética , Neoplasias/diagnóstico por imagen , Infecciones Estafilocócicas/diagnóstico por imagen , Agua/química , Acetatos/síntesis química , Acetatos/farmacocinética , Animales , Antibacterianos/uso terapéutico , Biopelículas , Ciprofloxacina/uso terapéutico , Portadores de Fármacos/síntesis química , Femenino , Colorantes Fluorescentes/química , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Liposomas/química , Masculino , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/fisiología , Compuestos de Piridinio/síntesis química , Compuestos de Piridinio/farmacocinética , Ratas Sprague-Dawley , Rodaminas/química , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/fisiopatología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/fisiología , Tuberculosis/diagnóstico por imagen , Tuberculosis/fisiopatología
16.
Adv Funct Mater ; 30(48)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34737689

RESUMEN

Internalization of Staphylococcus aureus by macrophages can inactivate bacterial killing mechanisms, allowing intracellular residence and dissemination of infection. Concurrently, these staphylococci can evade antibiotics that are frequently unable to pass mammalian cell membranes. A binary, amphiphilic conjugate composed of triclosan and ciprofloxacin is synthesized that self-assemble through micelle formation into antimicrobial nanoparticles (ANPs). These novel ANPs are stabilized through encapsulation in macrophage membranes, providing membrane-encapsulated, antimicrobial-conjugated NPs (Me-ANPs) with similar protein activity, Toll-like receptor expression and negative surface charge as their precursor murine macrophage/human monocyte cell lines. The combination of Toll-like receptors and negative surface charge allows uptake of Me-ANPs by infected macrophages/monocytes through positively charged, lysozyme-rich membrane scars created during staphylococcal engulfment. Me-ANPs are not engulfed by more negatively charged sterile cells possessing less lysozyme at their surface. The Me-ANPs kill staphylococci internalized in macrophages in vitro. Me-ANPs likewise kill staphylococci more effectively than ANPs without membrane-encapsulation or clinically used ciprofloxacin in a mouse peritoneal infection model. Similarly, organ infections in mice created by dissemination of infected macrophages through circulation in the blood are better eradicated by Me-ANPs than by ciprofloxacin. These unique antimicrobial properties of macrophage-monocyte Me-ANPs provide a promising direction for human clinical application to combat persistent infections.

17.
Chem Soc Rev ; 48(2): 428-446, 2019 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-30601473

RESUMEN

Bacterial-infections are mostly due to bacteria in an adhering, biofilm-mode of growth and not due to planktonically growing, suspended-bacteria. Biofilm-bacteria are much more recalcitrant to conventional antimicrobials than planktonic-bacteria due to (1) emergence of new properties of biofilm-bacteria that cannot be predicted on the basis of planktonic properties, (2) low penetration and accumulation of antimicrobials in a biofilm, (3) disabling of antimicrobials due to acidic and anaerobic conditions prevailing in a biofilm, and (4) enzymatic modification or inactivation of antimicrobials by biofilm inhabitants. In recent years, new nanotechnology-based antimicrobials have been designed to kill planktonic, antibiotic-resistant bacteria, but additional requirements rather than the mere killing of suspended bacteria must be met to combat biofilm-infections. The requirements and merits of nanotechnology-based antimicrobials for the control of biofilm-infection form the focus of this Tutorial Review.


Asunto(s)
Antiinfecciosos/farmacología , Biopelículas/efectos de los fármacos , Portadores de Fármacos/química , Nanotecnología/métodos , Antiinfecciosos/química , Antiinfecciosos/uso terapéutico , Infecciones Bacterianas/tratamiento farmacológico , Farmacorresistencia Bacteriana/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/fisiología , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/fisiología , Humanos , Nanopartículas/química
18.
Nano Lett ; 19(11): 7662-7672, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31593471

RESUMEN

Tumor heterogeneity has been one of the most important factors leading to the failure of conventional cancer therapies due to the accumulation of genetically distinct tumor-cell subpopulations during the tumor development process. Due to the diversity of genetic mutations during tumor growth, combining the use of multiple drugs has only achieved limited success in combating heterogeneous tumors. Herein, we report a novel antitumor strategy that effectively addresses tumor heterogeneity by using a CRISPR/Cas9-based nanoRNP carrying a combination of sgRNAs. Such nanoRNP is synthesized from Cas9 ribonucleoprotein, any combinations of required sgRNAs, and a rationally designed responsive polymer that endows nanoRNP with high circulating stability, enhanced tumor accumulation, and the efficient gene editing in targeted tumor cells eventually. By carrying a combination of sgRNAs that targets STAT3 and RUNX1, the nanoRNP exhibited efficient gene expression disruptions on a heterogeneous tumor model with two subsets of cells whose proliferations were sensitive to the reduced expression of STAT3 and RUNX1, respectively, leading to the effective growth inhibition of the heterogeneous tumor. Considering the close relationship between tumor heterogeneity and cancer progression, resistance to therapy, and recurrences, nanoRNP provides a feasible strategy to overcome tumor heterogeneity in the development of more advanced cancer therapy against malignant tumors.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Neoplasias/terapia , Animales , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Terapia Genética/métodos , Humanos , Ratones , Ratones Desnudos , Nanomedicina/métodos , Neoplasias/genética , Neoplasias/patología , Factor de Transcripción STAT3/genética
19.
Nano Lett ; 19(2): 674-683, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30444372

RESUMEN

Alzheimer's disease (AD) is a progressive and irreversible brain disorder. Recent studies revealed the pivotal role of ß-amyloid (Aß) in AD. However, there is no conclusive indication that the existing therapeutic strategies exerted any effect on the mitigation of Aß-induced neurotoxicity and the elimination of Aß aggregates simultaneously in vivo. Herein, we developed a novel nanocomposite that can eliminate toxic Aß aggregates and mitigate Aß-induced neurotoxicity in AD mice. This nanocomposite was designed to be a small-sized particle (14 ± 4 nm) with Aß-binding peptides (KLVFF) integrated on the surface. The nanocomposite was prepared by wrapping a protein molecule with a cross-linked KLVFF-containing polymer layer synthesized by in situ polymerization. The presence of the nanocomposite remarkably changed the morphology of Aß aggregates, which led to the formation of Aß/nanocomposite coassembled nanoclusters instead of Aß oligomers. With the reduction of the pathological Aß oligomers, the nanocomposites attenuated the Aß-induced neuron damages, regained endocranial microglia's capability to phagocytose Aß, and eventually protected hippocampal neurons against apoptosis. Thus, we anticipate that the small-sized nanocomposite will potentially offer a feasible strategy in the development of novel AD treatments.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Nanocompuestos/uso terapéutico , Nanomedicina/métodos , Péptidos/uso terapéutico , Agregación Patológica de Proteínas/terapia , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/aislamiento & purificación , Animales , Modelos Animales de Enfermedad , Ratones , Modelos Moleculares , Nanocompuestos/química , Nanocompuestos/ultraestructura , Péptidos/química , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología
20.
Angew Chem Int Ed Engl ; 59(18): 7235-7239, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32061182

RESUMEN

While poly(acyclic orthoester)s (PAOEs) have many appealing features for drug delivery, their application is significantly hindered by a lack of facile synthetic methods. Reported here is a simple method for synthesizing acyclic diketene acetal monomers from diols and vinyl ether, and their polymerization with a diol to first synthesize PAOEs. The PAOEs rapidly hydrolyze at lysosomal pH. With the help of a cationic lipid, ovalbumin, a model vaccine antigen was efficiently loaded into PAOEs nanoparticles using a double emulsion method. These nanoparticles efficiently delivered ovalbumin into the cytosol of dendritic cells and demonstrated enhanced antigen presentation over poly(lactic-co-glycolic acid) (PLGA) nanoparticles. PAOEs are promising vehicles for intracellular delivery of biopharmaceuticals and could increase the utility of poly(orthoesters) in biomedical research.


Asunto(s)
Materiales Biocompatibles/síntesis química , Ovalbúmina/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/inmunología , Vacunas/inmunología , Presentación de Antígeno/inmunología , Materiales Biocompatibles/química , Citosol/química , Citosol/inmunología , Estructura Molecular , Nanopartículas/química , Ovalbúmina/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/síntesis química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Vacunas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA