Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Commun ; 15(1): 328, 2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38184609

RESUMEN

Membrane-camouflaged nanomedicines often suffer from reduced efficacy caused by membrane protein disintegration and spatial disorder caused by separation and reassembly of membrane fragments during the coating process. Here we show that intracellularly gelated macrophages (GMs) preserve cell membrane structures, including protein content, integration and fluidity, as well as the membrane lipid order. Consequently, in our testing GMs act as cellular sponges to efficiently neutralize various inflammatory cytokines via receptor-ligand interactions, and serve as immune cell-like carriers to selectively bind inflammatory cells in culture medium, even under a flow condition. In a rat model of collagen-induced arthritis, GMs alleviate the joint injury, and suppress the overall arthritis severity. Upon intravenous injection, GMs efficiently accumulate in the inflammatory lungs of acute pneumonia mice for anti-inflammatory therapy. Conveniently, GMs are amenable to lyophilization and can be stored at ambient temperatures for at least 1 month without loss of integrity and bio-activity. This intracellular gelation technology provides a universal platform for targeted inflammation neutralization treatment.


Asunto(s)
Artritis Experimental , Ratas , Ratones , Animales , Artritis Experimental/tratamiento farmacológico , Medios de Cultivo , Citocinas , Liofilización , Macrófagos
2.
J Control Release ; 360: 82-92, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37331605

RESUMEN

Camptothecin (CPT) and cisplatin (Pt) have shown synergistic effects on a variety of cancers during preclinical and clinical studies. However, the ratio of the two drugs often could not be precisely regulated in different delivery systems, which hinders the desired synergistic effect. In addition, the low delivery efficiency of the two drugs to the tumor further impedes the ideal therapeutic outcomes. Herein, we report that a platelet-mimicking supramolecular nanomedicine (SN) could precisely control of the ratio of CPT and Pt with a high tumor accumulation rate for cascade amplification of synergistic chemotherapy. The SN was fabricated via the host-guest interaction between cucurbit[7]uril conjugated hyaluronic acid (HA-CB[7]) and adamantane (ADA) respectively functionalized CPT- and Pt-based prodrugs. The ratio of CPT and Pt in the SN could be facilely regulated by simply controlling the loading ratio, based on the strong binding affinity between CB[7] and ADA, and SN60 with 60% CPT and 40% Pt showed the highest synergistic effects on 4T1 cells. To improve the tumor accumulation efficiency of SN, 5,6-dimethylxanthenone-4-acetic acid (DMXAA, a tumor vasculature-disruptive agent) was loaded into the optimized SN and then coated with platelet membrane to yield platelet-mimicking supramolecular nanomedicine (D@SN-P). D@SN-P could first passively accumulate in tumors owing to the enhanced permeability and retention (EPR) effect after intravenous administration. The initially release of DMXAA from D@SN-P could induce tumor vascular disruption, and the resultant epithelial collagen exposure around the disrupted tumor vasculature provided a target for further recruitment of platelet-mimicking SN, leading to cascade amplification of tumor accumulation with synergistic chemotherapy. Hence, this platelet-mimicking supramolecular nanomedicine presents a universal supramolecular strategy to finely regulate the ratio of loaded pro-drugs, and improve the accumulation efficiency to amplify chemotherapy via platelet-mimics.


Asunto(s)
Neoplasias , Profármacos , Humanos , Camptotecina , Nanomedicina , Neoplasias/tratamiento farmacológico , Cisplatino/uso terapéutico , Línea Celular Tumoral
3.
ACS Appl Mater Interfaces ; 15(24): 29012-29022, 2023 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-37291057

RESUMEN

A cell-based drug delivery system has emerged as a promising drug delivery platform. Due to their innate inflammatory tropism, natural and engineered macrophages have exhibited targeted accumulation in inflammatory tissues, which has allowed targeted delivery of medicine for the treatment of a variety of inflammatory diseases. Nevertheless, live macrophages may take up the medicine and metabolize it during preparation, storage, and in vivo delivery, sometimes causing unsatisfactory therapeutic efficacy. In addition, live macrophage-based drug delivery systems are usually freshly prepared and injected, due to the poor stability that does not allow storage. "Off-the-shelf" products would be indeed conducive to the timely therapy of acute diseases. Herein, a cryo-shocked macrophage-based drug delivery system was developed via supramolecular conjugation of cyclodextrin (CD)-modified "zombie" macrophages and adamantane (ADA)-functionalized nanomedicine. "Zombie" macrophages exhibited a much better storage stability over time than their counterpart live macrophage drug carriers and maintained cell morphology, membrane integrity, and biological functions. In an acute pneumonia mouse model, "zombie" macrophages carried quercetin-loaded nanomedicine, hand-in-hand, to the inflammatory lung tissues and effectively alleviated the inflammation in mice.


Asunto(s)
Ciclodextrinas , Neumonía , Animales , Ratones , Sistemas de Liberación de Medicamentos , Portadores de Fármacos/farmacología , Macrófagos , Ciclodextrinas/farmacología
4.
ACS Appl Bio Mater ; 6(9): 3463-3471, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37161309

RESUMEN

UV exposure often triggers photoaging of the skin. Pharmacological treatment suffers from severe side effects as well as poor efficacy because of insufficient skin penetration. Dissolved oxygen has been previously shown to reverse photoaged skin; however, the treatment is often limited by the availability of equipment (e.g., high-pressure oxygen). Poor oxygen diffusion into the skin has also limited its therapeutic efficacy. Herein, we developed a microneedle patch to deliver living microalgae to the deeper layers of the skin for efficient oxygenation and reversal of photoaging. The continuous release of oxygen from microalgae in the skin through photosynthesis reversed the inflammatory microenvironment and reduced reactive oxygen species levels in the photodamaged skin, leading to collagen regeneration and reduced wrinkles. This study provides not only a means for highly efficient skin oxygenation and reversal of photoaging but also an important theoretical basis for the clinical treatment of photoaging.


Asunto(s)
Microalgas , Envejecimiento de la Piel , Rayos Ultravioleta/efectos adversos , Oxígeno/farmacología , Piel
5.
ACS Nano ; 17(4): 4034-4049, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36739531

RESUMEN

Sonodynamic therapy (SDT) is a noninvasive technique for local antitumor treatment; however, its clinical application is often limited by the low tumor accumulation of SDT agents, tumor's hypoxic microenvironment, and cytoprotective effects of autophagy. To address these issues, herein we developed surface-engineered chlorella (Chl, a green algae) as a targeted drug carrier and sustainable oxygen supplier (via photosynthesis) for significantly improved SDT via hypoxia alleviation as well as autophagy inhibition of chloroquine phosphate. In this design, the macrophage membrane was coated onto Chl to form macrophage-mimetic Chl (MChl) to increase its biocompatibility and targeted tumor accumulation driven by the inflammatory-homing effects of macrophage membranes. In addition, the membrane coating on Chl allowed lipid insertion to yield ß-cyclodextrin (ß-CD) modified MChl (CD-MChl). Subsequently, supramolecular conjugates of MChl-NP were constructed via host-guest interactions between CD-MChl and adamantane (ADA)-modified liposome (ADA-NP), and the anchored liposome went with CD-MChl hand-in-hand to the tumor tissues for co-delivery of Chl, hematoporphyrin, and chloroquine phosphate (loaded in ADA-NP). The synergistic therapy achieved via local oxygenation, SDT, and autophagy inhibition maximally improved the therapeutic efficacy of MChl-CQ-HP-NP against melanoma. Tumor rechallenging results revealed that the changes of tumor microenvironment including hypoxia alleviation, SDT induced immunogenic cell death, and autophagy inhibition collectively induced a strong antitumor immune response and memory.


Asunto(s)
Chlorella , Microalgas , Terapia por Ultrasonido , Humanos , Liposomas/farmacología , Línea Celular Tumoral , Chlorella/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Hipoxia/metabolismo , Inmunoterapia , Autofagia , Macrófagos/metabolismo , Terapia por Ultrasonido/métodos
6.
J Control Release ; 349: 2-15, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35779655

RESUMEN

Current pharmacological treatments of atherosclerosis often target either cholesterol control or inflammation management, to inhibit atherosclerotic progression, but cannot lead to direct plaque lysis and atherosclerotic regression, partly due to the poor accumulation of medicine in the atherosclerotic plaques. Due to enhanced macrophage recruitment during atheromatous plaque progression, a macrophage-liposome conjugate was facilely constructed for targeted anti-atherosclerosis therapy via synergistic plaque lysis and inflammation alleviation. Endogenous macrophage is utilized as drug-transporting cell, upon membrane-modification with a ß-cyclodextrin (ß-CD) derivative to form ß-CD decorated macrophage (CD-MP). Adamantane (ADA) modified quercetin (QT)-loaded liposome (QT-NP), can be conjugated to CD-MP via host-guest interactions between ß-CD and ADA to form macrophage-liposome conjugate (MP-QT-NP). Thus, macrophage carries liposome "hand-in-hand" to significantly increase the accumulation of anchored QT-NP in the aorta plaque in response to the plaque inflammation. In addition to anti-inflammation effects of QT, MP-QT-NP efficiently regresses atherosclerotic plaques from both murine aorta and human carotid arteries via CD-MP mediated cholesterol efflux, due to the binding of cholesterol by excess membrane ß-CD. Transcriptome analysis of atherosclerotic murine aorta and human carotid tissues reveal that MP-QT-NP may activate NRF2 pathway to inhibit plaque inflammation, and simultaneously upregulate liver X receptor to promote cholesterol efflux.


Asunto(s)
Adamantano , Aterosclerosis , Ciclodextrinas , Placa Aterosclerótica , beta-Ciclodextrinas , Adamantano/metabolismo , Adamantano/farmacología , Adamantano/uso terapéutico , Animales , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Colesterol/metabolismo , Ciclodextrinas/farmacología , Humanos , Inflamación/metabolismo , Liposomas/metabolismo , Receptores X del Hígado , Macrófagos , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/farmacología , Factor 2 Relacionado con NF-E2/uso terapéutico , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/metabolismo , Quercetina/farmacología , Quercetina/uso terapéutico , beta-Ciclodextrinas/uso terapéutico
7.
Sci Adv ; 8(19): eabn1805, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35544569

RESUMEN

Cell-based drug carriers are mostly prepared in vitro, which may negatively affect the physiological functions of cells, and induce possible immune rejections when applied to different individuals. In addition, the immunosuppressive tumor microenvironment limits immune cell-mediated delivery. Here, we report an in vivo strategy to construct cell-based nanomedicine carriers, where bacteria-mimetic gold nanoparticles (GNPs) are intravenously injected, selectively phagocytosed by phagocytic immune cells, and subsequently self-assemble into sizable intracellular aggregates via host-guest interactions. The intracellular aggregates minimize exocytosis of GNPs from immune cells and activate the photothermal property via plasmonic coupling effects. Phagocytic immune cells carry the intracellular GNP aggregates to melanoma tissue via inflammatory tropism. Moreover, an initial photothermal treatment (PTT) of the tumor induces tumor damage that subsequently provides positive feedback to recruit more immune cell-based carriers for enhanced targeting efficiency. The optimized secondary PTT notably improves antitumor immunotherapy, further strengthened by immune checkpoint blockade.


Asunto(s)
Melanoma , Nanopartículas del Metal , Nanopartículas , Neoplasias , Bacterias , Línea Celular Tumoral , Oro , Humanos , Melanoma/tratamiento farmacológico , Nanomedicina , Microambiente Tumoral
8.
Biomater Sci ; 9(10): 3804-3813, 2021 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-33881050

RESUMEN

Nano-graphene oxide (NGO) has attracted increasing attention as an advanced drug delivery system. However, the current surface functionalization and drug-loading of NGO either rely on π-π stacking that is limited to aromatic molecules, or covalent conjugation that requires tedious synthesis. Herein, we developed the first cucurbit[7]uril (CB[7])-conjugated NGO (NGO-CB[7]) that allows non-covalent, modular surface functionalization and drug loading via not only traditional π-π stacking interactions between the NGO surface and functional molecules, but also strong host-guest interactions between CB[7] and guest payloads or adamantane (ADA)-tagged functional molecules, for more versatile biomedical applications. To this end, chlorin e6 (Ce6, a photosensitizer), banoxantrone dihydrochloride (AQ4N, a hypoxia-responsive prodrug) and oxaliplatin (OX, a guest of CB[7]) were co-loaded onto NGO-CB[7] via π-π stacking and host-guest interactions, respectively. Subsequently, ADA-tagged hyaluronic acid (ADA-HA) wrapped NGO-CB[7] non-covalently via CB[7]-ADA host-guest interactions to improve the physiological stability and overall biocompatibility of this supramolecular nanosystem, and to enable targeted delivery into cancer cells with CD44 receptors overexpressed. Remarkably, this supramolecular nanomedicine exhibited significant antitumor efficacy via combined photothermal/photodynamic therapy (PTT/PDT) from NGO/Ce6, as well as dual chemotherapy from OX and AQ4N (activated by PDT-enhanced hypoxia), in vitro and in vivo. This study not only offers a new supramolecular inorganic/organic hybrid nanosystem for multi-modality cancer therapy, but may also provide important new insights into noncovalent functionalization of other carbon nanomaterials and inorganic nanomaterials leading to multifunctional drug delivery systems.


Asunto(s)
Nanomedicina , Neoplasias , Hidrocarburos Aromáticos con Puentes , Imidazoles , Neoplasias/tratamiento farmacológico , Óxidos
9.
Chin Chem Lett ; 32(10): 3019-3022, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33840982

RESUMEN

The wide-spreading SARS-CoV-2 virus has put the world into boiling water for more than a year, however pharmacological therapies to act effectively against coronavirus disease 2019 (COVID-19) remain elusive. Chloroquine (CQ), an antimalarial drug, was found to exhibit promising antiviral activity in vitro and in vivo at a high dosage, thus CQ was approved by the FDA for the emergency use authorization (EUA) in the fight against COVID-19 in the US, but later was revoked the EUA status due to the severe clinical toxicity. Herein, we show that supramolecular formulation of CQ by a macrocyclic host, curcurbit[7]uril (CB[7]), reduced its non-specific toxicity and improved its antiviral activity against coronavirus, working in synergy with CB[7]. CB[7] was found to form 1:1 host-guest complexes with CQ, with a binding constant of ∼104 L/mol. The CQ-CB[7] formulation decreased the cytotoxicity of CQ against Vero E6 and L-02 cell lines. In particular, the cytotoxicity of CQ (60 µmol/L) against both Vero E6 cell line and L-02 cell lines was completely inhibited in the presence of 300 µmol/L and 600 µmol/L CB[7], respectively. Furthermore, the CB[7] alone showed astonishing antiviral activity in SARS-CoV-2 infected Vero E6 cells and mouse hepatitis virus strain A59 (MHV-A59) infected N2A cells, and synergistically improved the antiviral activity of CQ-CB[7], suggesting that CB[7]-based CQ formulation has a great potential as a safe and effective antiviral agent against SARS-CoV-2 and other coronavirus.

10.
Theranostics ; 10(22): 10106-10119, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32929337

RESUMEN

Ulcerative colitis (UC) is featured with relapsing inflammation in the colon, where macrophages are recruited and polarized locally into M1 type to drive further inflammation. Pharmacotherapy of UC has exhibited limited efficacy, mostly due to the poor specificity. Methods: A macrophage-biomimetic nanomedicine was developed for targeted treatment of UC, which was derived from reactive oxygen species (ROS)-sensitive ß-cyclodextrin, loaded with rosiglitazone, and coated with macrophage membrane. The ability of the nanomedicine in regulating macrophage polarization was examined at cellular level, and the macrophage-tropism driven targeted delivery into the inflammatory colon was investigated by ex vivo bio-imaging distribution assay. Furthermore, the nanomedicine's therapeutic efficacy was systemically examined in dextran sulfate sodium (DSS)-induced colitis model in mice. Results: The nanomedicine effectively polarized macrophages to M2 and protected epithelial cells from oxidative stress in vitro. In addition, macrophage-membrane led the nanomedicine to the inflammatory colon with a high targeting efficiency. In response to the elevated ROS in the inflammatory tissue, the nanomedicine released rosiglitazone specifically and regulated macrophage polarization in vivo. Macrophage membrane also assisted inflammation suppression by sequestering proinflammatory cytokines. Working in such a synergy, the nanomedicine exhibited significant therapeutic effects against UC in mice. Conclusions: This macrophage-biomimetic nanomedicine leverages the inflammatory tropism and inflammatory cytokine sequestration effects of macrophage membrane for targeted delivery and local inflammation suppression, the ROS-responsiveness of ß-cyclodextrin-based matrix for specific payload release, and the macrophage-polarizing effect of rosiglitazone for inflammatory regulation, thereby exhibiting considerable therapeutic efficacy against UC in mice. This study offers important new insights on the design and development of biomimetic nanomaterials for inflammation regulations.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Animales , Biomimética/métodos , Células CACO-2 , Línea Celular , Línea Celular Tumoral , Colitis/tratamiento farmacológico , Colitis/metabolismo , Colitis Ulcerosa/metabolismo , Colon/efectos de los fármacos , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Nanomedicina/métodos , Estrés Oxidativo/efectos de los fármacos , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo
11.
ACS Appl Mater Interfaces ; 12(23): 25604-25615, 2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-32406668

RESUMEN

In spite of the rapid emergence of numerous nanoparticles (NPs) for biomedical applications, it is often challenging to precisely control, or effectively tame, the bioactivity/toxicity of NPs, thereby exhibiting limited applications in biomedical areas. Herein, we report the construction of hyaluronic acid (HA)-laminated, otherwise toxic methylviologen (MV), NPs via ternary host-guest complexation among cucurbit[8]uril, trans-azobenzene-conjugated HA, and MV-functionalized polylactic acid NPs (MV-NPs). The high, nonspecific toxicity of MV-NPs was effectively shielded (turned off) by HA lamination, as demonstrated in cells, zebrafish, and mouse models. The supramolecular host-guest interaction-mediated HA coating offered several HA-MV-NP modalities, including hyaluronidase locally and photoirradiation remotely, to precisely remove HA lamination on demand, thereby endowing materials with the capability of selective decoating-induced activation (DIA) for applications as a user-friendly herbicide, a selective antibacterial agent, or an anticancer nanomedicine. This work offers facile supramolecular coating and DIA strategies to effectively tame and precisely control the bioactivity and toxicity of functional nanomaterials for diverse applications.


Asunto(s)
Antibacterianos/uso terapéutico , Antineoplásicos/uso terapéutico , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Paraquat/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Animales , Antibacterianos/química , Antibacterianos/toxicidad , Antineoplásicos/química , Antineoplásicos/toxicidad , Hidrocarburos Aromáticos con Puentes/química , Hidrocarburos Aromáticos con Puentes/toxicidad , Línea Celular Tumoral , Escherichia coli/efectos de los fármacos , Femenino , Fluoruros/química , Fluoruros/efectos de la radiación , Gadolinio/química , Gadolinio/efectos de la radiación , Ácido Hialurónico/química , Ácido Hialurónico/toxicidad , Imidazoles/química , Imidazoles/toxicidad , Rayos Infrarrojos , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Nanopartículas/química , Nanopartículas/efectos de la radiación , Nanopartículas/toxicidad , Paraquat/química , Paraquat/toxicidad , Poliésteres/química , Poliésteres/toxicidad , Staphylococcus aureus/efectos de los fármacos , Tulio/química , Tulio/efectos de la radiación , Iterbio/química , Iterbio/efectos de la radiación , Pez Cebra
12.
Nat Commun ; 11(1): 2622, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32457361

RESUMEN

Vascular disease remains the leading cause of death and disability, the etiology of which often involves atherosclerosis. The current treatment of atherosclerosis by pharmacotherapy has limited therapeutic efficacy. Here we report a biomimetic drug delivery system derived from macrophage membrane coated ROS-responsive nanoparticles (NPs). The macrophage membrane not only avoids the clearance of NPs from the reticuloendothelial system, but also leads NPs to the inflammatory tissues, where the ROS-responsiveness of NPs enables specific payload release. Moreover, the macrophage membrane sequesters proinflammatory cytokines to suppress local inflammation. The synergistic effects of pharmacotherapy and inflammatory cytokines sequestration from such a biomimetic drug delivery system lead to improved therapeutic efficacy in atherosclerosis. Comparison to macrophage internalized with ROS-responsive NPs, as a live-cell based drug delivery system for treatment of atherosclerosis, suggests that cell membrane coated drug delivery approach is likely more suitable for dealing with an inflammatory disease than the live-cell approach.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Citocinas/metabolismo , Sistemas de Liberación de Medicamentos , Macrófagos/metabolismo , Nanopartículas/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aterosclerosis/metabolismo , Atorvastatina/uso terapéutico , Materiales Biomiméticos , Membrana Celular/metabolismo , Liberación de Fármacos , Femenino , Ratones , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Resultado del Tratamiento
13.
J Mater Chem B ; 8(14): 2749-2753, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32215428

RESUMEN

Herein we developed cucurbit[7]uril covalently modified Fe3O4 nanoparticles for facile surface modification via host-guest interactions to realize targeted drug delivery and magnetic resonance imaging of tumors in vivo.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/química , Imidazoles/química , Nanopartículas de Magnetita/química , Neoplasias/diagnóstico por imagen , Animales , Sistemas de Liberación de Medicamentos , Humanos , Imagen por Resonancia Magnética , Neoplasias/tratamiento farmacológico , Tamaño de la Partícula , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA