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.
J Transl Med ; 22(1): 78, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38243324

RESUMEN

BACKGROUND: Inflammatory Bowel Diseases (IBD), an autoimmune disease characterised by abnormal intestinal immunity, are related to vital morbidity around the world. However, therapeutic agents for IBD have not achieved desired benefit. Exploring new therapeutic targets for IBD, especially based on its abnormally intestinal immunity, could alleviate the flare-up and worsening of IBD. Tissue resident memory T cells (TRM) are core of multiple autoimmune diseases, including IBD. However, the mechanism of TRM differentiation remains to be investigated. METHODS: The alterations in mRNA and lncRNA profile of intestinal intraepithelial lymphocytes (IELs), the largest component of intestinal TRM, were analyzed in DSS-induced chronic colitis. Based on it, we examined the function of rectal insulin instillation in a dextran sodium sulfate (DSS) induced chronic colitis. Furthermore, we investigated the downstream-target of the insulin pathway-EZH2 and the crucial role of EZH2 in intestinal tissue resident memory T cell differentiation by utilizing EZH2fl/flCD4cre mice. RESULTS: Insulin receptor (INSR) expression was found to be significantly reduced. Activation of mucosal insulin pathway by rectal insulin instillation exacerbated colitis by disrupting IELs subgroups and up-regulating TNF-ɑ and IL-17 expression. Rectal insulin instillation promoted EZH2 expression and EZH2 inhibition alleviated chronic colitis. EZH2fl/flCD4cre mice restored the normal IEL subgroups and suppressed TNF-ɑ and IL-17 expression, exhibiting alleviated colitis. IELs from EZH2fl/flCD4cre mice exhibit significant changes in TRM related phenotype. CD4+TRM was significantly increased in chronic colitis and decreased in EZH2fl/flCD4cre mice. CONCLUSION: Insulin receptor of intestinal mucosal T-cells could promote intestinal TRM differentiation via EZH2. Our discoveries suggest that therapies targeting colonic INSR and EZH2 could be potential treatment for IBD based on its regulatory effects on TRM. Insulin receptor inhibitors rather than insulin should be applied during colitis-active phase. In addition, EZH2 shows to be a downstream signal of the insulin pathway and EZH2 inhibitor could alleviating intestinal inflammation. However, the critical role of EZH2 in TRM differentiation restricts the anti-tumor effects of EZH2 inhibitor in vivo.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Insulinas , Ratones , Animales , Interleucina-17/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptor de Insulina/efectos adversos , Receptor de Insulina/metabolismo , Células T de Memoria , Colitis/inducido químicamente , Diferenciación Celular , Mucosa Intestinal/patología , Inflamación/patología , Insulinas/metabolismo , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad
2.
J Mater Chem B ; 11(43): 10369-10382, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37873599

RESUMEN

Ceria nanozyme-based ROS scavengers have shown great potential in the treatment of inflammatory bowel disease (IBD) through microenvironment regulation. However, the currently developed nanotherapeutics suffer from difficulties in concomitantly achieving small sizes and stable interparticle dispersion which is pivotal to sufficient oxygen vacancies facilitating electron transfer and oxygen storage in the dynamic cycling of Ce3+/Ce4+ redox pairs. Herein, a hybrid nanosystem consisting of ceria nanodots supported on redox-active mesoporous hosts was developed to address the challenge of ROS scavenging, in particular the efficient downregulation of the readily renewable, highly concentrated H2O2 species. Specifically, Ce4+ ions oxidized from Ce3+ in weakly basic solution were captured and reduced in time by the abundant catechols on the mesoporous polydopamine nanoparticles. This led to strong restriction of ceria growth (∼2.8 nm) in the ion precipitation process and efficient maintenance of the Ce3+/Ce4+ ratio at a high value of 1.59 which is 4.8 fold higher than that of homogeneously nucleated ceria nanoparticles. Through this design, the nanohybrid showed an attractive catalytic performance in scavenging multiple ROS species, particularly the fast and recyclable conversion of H2O2. Thereby, significant suppression of the inflammatory cytokine/chemokine secretion was achieved by inhibiting the activation of NF-κB signaling pathways (5.1 fold higher as compared to those of pristine ceria nanoparticles), upregulating the Nrf2 signaling pathway, and reducing the proportion of M1 macrophages at IBD sites. Therapeutic efficiency was also demonstrated by the effective repair of the intestinal mucosal barrier by recovering the tight junction integrity in vivo. This study sheds light on the employment of redox-active hosts to support ceria catalysts for advancing anti-inflammation applications by boosting ROS scavenging performance.


Asunto(s)
Peróxido de Hidrógeno , Enfermedades Inflamatorias del Intestino , Humanos , Especies Reactivas de Oxígeno/metabolismo , Oxidación-Reducción , Oxígeno , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico
3.
Clin Transl Med ; 13(7): e1321, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37400975

RESUMEN

BACKGROUND: Although ileal faecal diversion is commonly used in clinical settings, complications accompany it. Elucidating the intestinal changes caused by ileal faecal diversion will help resolve postoperative complications and elucidate the pathogenic mechanisms of associated intestinal disorders, such as Crohn's disease (CD). Therefore, our study aimed to provide new insights into the effects of ileal faecal diversion on the intestine and the potential mechanisms. METHODS: Single-cell RNA sequencing was performed on proximal functional and paired distal defunctioned intestinal mucosae from three patients with ileal faecal diversion. We also performed in vitro cellular and animal experiments, tissue staining and analysed public datasets to validate our findings. RESULTS: We found that the epithelium in the defunctioned intestine tended to be immature, with defective mechanical and mucous barriers. However, the innate immune barrier in the defunctioned intestine was enhanced. Focusing on the changes in goblet cells, we demonstrated that mechanical stimulation promotes the differentiation and maturation of goblet cells through the TRPA1-ERK pathway, indicating that the absence of mechanical stimulation may be the main cause of defects in the goblet cells of the defunctioned intestine. Furthermore, we found obvious fibrosis with a pro-fibrotic microenvironment in the defunctioned intestine and identified that monocytes may be important targets for faecal diversion to alleviate CD. CONCLUSIONS: This study revealed the different transcription landscapes of various cell subsets and the potential underlying mechanisms within the defunctioned intestine, when compared to the functional intestine, based on the background of ileal faecal diversion. These findings provide novel insights for understanding the physiological and pathological roles of the faecal stream in the intestine.


Asunto(s)
Enfermedad de Crohn , Ileostomía , Humanos , Ileostomía/efectos adversos , Enfermedad de Crohn/etiología , Enfermedad de Crohn/patología , Enfermedad de Crohn/cirugía , Heces , Complicaciones Posoperatorias/patología , Mucosa Intestinal/patología
4.
World J Gastroenterol ; 29(13): 1942-1954, 2023 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-37155525

RESUMEN

The tremendous public health and economic impact of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a huge challenge globally. There is increasing evidence that SARS-CoV-2 induces intestinal infections. Type III interferon (IFN-λ) has an antiviral role in intestinal infection, with focused, long-lasting, and non-inflammatory characteristics. This review presents a summary of the structure of SARS-CoV-2, including its invasion and immune escape mechanisms. Emphasis was placed on the gastrointestinal impact of SARS-CoV-2, including changes to the intestinal microbiome, activation of immune cells, and inflammatory responses. We also describe the comprehensive functions of IFN-λ in anti-enteric SARS-CoV-2 infection, and discuss the potential application of IFN-λ as a therapeutic agent for COVID-19 with intestinal symptoms.


Asunto(s)
COVID-19 , Humanos , Interferón lambda , SARS-CoV-2 , Interferones/uso terapéutico , Antivirales/uso terapéutico , Antivirales/farmacología
5.
Inflammation ; 46(1): 18-34, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36050591

RESUMEN

Tight junctions (TJs) are located in the apical region of the junctions between epithelial cells and are widely found in organs such as the brain, retina, intestinal epithelium, and endothelial system. As a mechanical barrier of the intestinal mucosa, TJs can not only maintain the integrity of intestinal epithelial cells but also maintain intestinal mucosal permeability by regulating the entry of ions and molecules into paracellular channels. Therefore, the formation disorder or integrity destruction of TJs can induce damage to the intestinal epithelial barrier, ultimately leading to the occurrence of various gastrointestinal diseases, such as inflammatory bowel disease (IBD), gastroesophageal reflux disease (GERD), and irritable bowel syndrome (IBS). However, a large number of studies have shown that TJs protein transport disorder from the endoplasmic reticulum to the apical membrane can lead to TJs formation disorder, in addition to disruption of TJs integrity caused by external pathological factors and reduction of TJs protein synthesis. In this review, we focus on the structural composition of TJs, the formation of clathrin-coated vesicles containing transmembrane TJs from the Golgi apparatus, and the transport process from the Golgi apparatus to the plasma membrane via microtubules and finally fusion with the plasma membrane. At present, the mechanism of the intracellular transport of TJ proteins remains unclear. More studies are needed in the future to focus on the sorting of TJs protein vesicles, regulation of transport processes, and recycling of TJ proteins, etc.


Asunto(s)
Intestinos , Proteínas de Uniones Estrechas , Proteínas de Uniones Estrechas/metabolismo , Mucosa Intestinal/metabolismo , Células Epiteliales/metabolismo , Uniones Estrechas/metabolismo
6.
ACS Appl Mater Interfaces ; 14(17): 19975-19987, 2022 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-35442639

RESUMEN

Benefiting from the evolution of nanotechnology, the combination therapy by gene interference and reactive oxygen species (ROS) scavenging are expected, which holds great potential in inflammatory bowel disease (IBD) therapy. However, the functional integration of different therapeutic modules through interface modification of gene vectors for safe and efficient treatment is urgently needed. Herein, we present a catechol chemistry-mediated core-shell nanoplatform for ROS scavenging-mediated oxidative stress alleviation and siRNA-mediated gene interference in a dextran sulfate sodium (DSS)-induced colitis model. The nanoplatform is constructed by employing mesoporous polydopamine nanoparticles (MPDA NPs) with surface modification of amines as the porous core for TNF-α-siRNA loading (31 wt %) and exerts an antioxidant function, while PDA-induced biomineralization of the calcium phosphate (CaP) coating is used as the pH-sensitive protective shell to prevent siRNA from premature release. The CaP layer degraded under weakly acidic subcellular conditions (lysosomes); thus, the synergistic integration of catechol and cation moieties on the exposed surface of MPDA resulted in an efficient lysosomal escape. Subsequently, effective ROS scavenging caused by the electron-donating ability of MPDA and efficient knocking down (40.5%) of tumor necrosis factor-α (TNF-α) via sufficient cytosolic gene delivery resulted in a synergistic anti-inflammation therapeutic effect both in vitro and in vivo. This work establishes the first paradigm of synergistic therapy in IBD by ROS scavenging and gene interference.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Nanopartículas , Catecoles/uso terapéutico , Humanos , Indoles , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/terapia , Polímeros , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Especies Reactivas de Oxígeno , Factor de Necrosis Tumoral alfa/genética
7.
Adv Healthc Mater ; 10(23): e2101476, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34599858

RESUMEN

The development of wound dressings with combined antibacterial activities and pro-healing functions has always been an intractable medical task for treating bacterial wound infection. Herein, a novel injectable hybrid hydrogel dressing is developed, which is doped with nitric oxide (NO) donor (N,N'-di-sec-butyl-N,N'-dinitroso-1,4-phenylenediamine, BNN6) loaded two-dimensional polydopamine nanosheets (PDA NS). The hydrogel matrix is in situ formed through dynamic Schiff base crosslinking between hydrazide-modified γ-polyglutamic acid (γ-PGA-ADH) and aldehyde-terminated Pluronic F127 (F127-CHO). Under 808 nm irradiation, the embedded PDA NS exhibits outstanding photothermal transform properties (56.1%) and on-demand NO release. The combination of photothermal and NO gas therapy with a synergistic antibacterial effect works on both Escherichia coli and Staphylococcus aureus in vitro. Furthermore, a full-thickness skin defect model also demonstrates that the hybrid hydrogel shows outstanding antibacterial properties and effectively accelerates the wound healing process. Overall, this study provides a facile and promising method for the fabrication of PDA NS based multifunctional hydrogel dressing for the application of infectious skin wound healing.


Asunto(s)
Hidrogeles , Óxido Nítrico , Antibacterianos/farmacología , Indoles , Polímeros , Cicatrización de Heridas
8.
J Inflamm Res ; 14: 1701-1716, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33953597

RESUMEN

The effective colon drug delivery remains to be an international frontier research in inflammatory bowel disease (IBD) therapy. The exploration and research of nanocarrier-based nanomedicine with great potential brings new opportunities for IBD therapy and diagnoses. Functional nanocarriers with varying morphology and characteristics can not only effectively avoid the destruction of the complex gastrointestinal (GI) tract microenvironment but also endow drugs with target therapy and improved bioavailability, thus elevating therapeutic efficacy. In this review, we illustrated several challenges in IBD therapy, then emphasis on some latest research progress of nanoparticles based therapy of oral administration, rectal administration and parenteral administration, as well as IBD diagnoses. Finally, we described the future perspective of nanocarriers in the treatment and diagnoses of IBD.

9.
Adv Healthc Mater ; 9(20): e2000432, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32945146

RESUMEN

Inhibiting the function of P-glycoprotein (P-gp) transporter, which causes drug efflux through adenosine triphosphate (ATP)-dependent manner, has become an effective strategy to conquer multidrug resistance (MDR) of cancer cells. However, there remains challenges for effective co-delivery, sequential release of P-gp modulator and chemotherapeutic agent. In this work, a novel type of core-shell nanoparticle is reported. It can independently encapsulate a high amount (about 683 µg mg-1 ) of chemotherapeutic agent doxorubicin (DOX) in the mesoporous polydopamine (MPDA) core and glucose oxidase (GOx) in the zeolite imidazolate frameworks-8 (ZIF-8) shell, namely MPDA@ZIF-8/DOX+GOx. The fast release of GOx triggered by acid-sensitive degradation of the ZIF-8 shell consumes glucose to starve cancer cells for ATP deprivation and effective suppress ATP-dependent drug efflux in advance, and then effectively facilitates the accumulation of DOX in MCF-7/ADR cancer cells. Experiments in vitro and in vivo demonstrate that the fabricated nanosystem can dramatically improve anticancer effects for MDR through sequential release property and exhibit excellent biocompatibility. Overall, this work reveals new insights in the use of GOx for MDR treatment.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Adenosina Trifosfato , Doxorrubicina/farmacología , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Humanos , Células MCF-7
10.
J Mol Med (Berl) ; 98(8): 1189-1202, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32696223

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is a critical regulator of barrier integrity during colonic mucosal injury. Previous works have shown that the absence of autophagy is implicated in the development of inflammatory bowel disease (IBD). Additionally, changes in bacterial profiles in the gut are intimately associated with IBD. Although HIF-1α, autophagy, microbiota, and their metabolites are all involved in the pathogenesis of IBD, their roles are not known. In this study, we investigated the relationship between HIF-1α and autophagy in healthy and inflammatory states using transgenic mice, colitis models, and cell culture models. We confirmed that the absence of intestinal epithelial HIF-1α changed the composition of the intestinal microbes and increased the susceptibility of mice to dextran sodium sulfate (DSS)-induced colitis. In addition, autophagy levels in the intestinal epithelial cells (IECs) were significantly reduced in IEC-specific HIF-1α-deficient (HIF-1α∆IEC) mice. Moreover, in the cell culture models, butyrate treatment significantly increased autophagy in HT29 cells under normal conditions, whereas butyrate had little effect on autophagy after HIF-1α ablation. Furthermore, in the DSS-induced colitis model, butyrate administration relieved the colonic injury and suppressed inflammation in Cre-/HIF-1α- (HIF-1αloxP/loxP) mice. However, the butyrate-mediated protection against colonic injury was considerably diminished in the HIF-1α∆IEC mice. These results show that HIF-1α, autophagy, and intestinal microbes are essential for the maintenance of intestinal homeostasis. Butyrate can alleviate DSS-induced colitis by regulating autophagy via HIF-1α. These insights may have important implications for the development of therapeutic strategies for IBD. KEY MESSAGES: • The absence of intestinal epithelial HIF-1α leads to downregulation of autophagy in mice. • The absence of intestinal epithelial HIF-1α exacerbates DSS-induced colitis. • Short-chain fatty acids (SCFAs) can alleviate DSS-induced colitis by regulating autophagy via HIF-1α.


Asunto(s)
Autofagia , Colitis/metabolismo , Ácidos Grasos Volátiles/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mucosa Intestinal/metabolismo , Animales , Autofagia/efectos de los fármacos , Biomarcadores , Colitis/etiología , Colitis/patología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Células Epiteliales/metabolismo , Ácidos Grasos Volátiles/farmacología , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Ratones
11.
ACS Appl Mater Interfaces ; 11(15): 13945-13953, 2019 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-30907570

RESUMEN

Currently, the simple integration of multiple therapeutic agents within a single nanostructure for combating multidrug resistance (MDR) tumors yet remains a challenge. Herein, we report a photoresponsive nanocluster (NC) system prepared by installing polydopamine (PDA) nanoparticle clusters on the surface of d-α-tocopheryl poly(ethylene glycol) 1000 succinate (TPGS) (a drug efflux inhibitor) micelles solubilized with IR780 (a photosensitizer) to achieve a combined chemotherapy (CT)/photothermal therapy (PTT)/photodynamic therapy (PDT) for drug-resistant breast cancer. Mediated by the fluorescence resonance energy transfer and radical scavenging properties of PDA, NC shows prominently quenched fluorescence emission (∼78%) and inhibited singlet oxygen generation (∼67%) upon exposure to near-infrared (NIR) light (808 nm, 0.5 W cm-2), favoring a highly efficient PTT module. Meanwhile, the photothermal heat can also boost the release of doxorubicin hydrochloride whose intracellular accumulation can be greatly enhanced by TPGS. Interestingly, the first NIR irradiation and subsequent incubation (∼24 h) can induce the gradual relocation and disintegration of PDA nanoparticles, thereby leading to activated PDT therapy under the second irradiation. Upon the temporally controlled sequential application of PTT/PDT, the developed NC exhibited a great potential to treat MDR cancer both in vitro and in vivo. These findings suggest that complementary interactions among PTT/PDT/CT modalities can enhance the efficiency of the combined therapy for MDR tumor.


Asunto(s)
Doxorrubicina/química , Indoles/química , Micelas , Nanopartículas/química , Polímeros/química , Animales , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Rayos Infrarrojos , Ratones , Ratones Desnudos , Nanopartículas/metabolismo , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Fototerapia , Polietilenglicoles/química , Especies Reactivas de Oxígeno/metabolismo , Trasplante Heterólogo
12.
Acta Biomater ; 86: 416-428, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30611792

RESUMEN

Combined photothermal and gene therapy provides a promising modality toward cancer treatment, yet facile integration and controlled codelivery of gene payloads and photothermal conversion agents (PTCAs) remains a great challenge. Inspired by the robust wet adhesion of marine mussels, we present a rationally designed nanosystem constructed by using hybrid mesoporous polydopamine nanoparticles (MPDA) with sub-100 nm sizes and a high photothermal conversion efficiency of 37%. The surface of the particles were modified with tertiary amines by the facile Michael addition/Schiff base reactions of PDA to realize high siRNA loading capacity (10 wt%). Moreover, a successful calcium phosphate (CaP) coating via biomineralization was constructed on the cationic nanoparticle to prohibit premature release of siRNA. The CaP coating underwent biodegradation in weakly-acidic subcellular conditions (lysosomes). The synergistic integration of tertiary amines and catechol moieties on the subsequently exposed surfaces was demonstrated to feature the destabilization/disruption ability toward model cellular membranes via the greatly enhanced interfacial adhesion and interactions. Consequently, sufficient permeability of lysosomal membranes, and in turn, a high lysosomal escape efficiency, was realized, which then resulted in high gene silencing efficiencies via sufficient cytosolic delivery of siRNA. When an efficient knocking down (65%) of survivin (an inhibitor of apoptosis proteins) was combined with a subsequent photothermal ablation, remarkably higher therapeutic efficiencies were observed both in vitro and in vivo, as compared with monotherapy. The system may help to pave a new avenue on the utilization of bio-adhesive surfaces for handling the obstacles of combined photothermal and gene therapy. STATEMENT OF SIGNIFICANCE: Polydopamine (PDA) based porous photothermal-conversion agent (PTCA) with sufficiently high conversion efficiency was employed to deliver photothermal/gene therapy modalities towards cancer treatment. CaP coating via PDA-induced biomineralization was constructed to prohibit premature release of siRNA loaded in the pore space of the nanocarriers. Responsive degradation of CaP also led to the exposure of membrane-lytic surfaces built through the synergistic integration of tertiary amines and catechol moieties, and in turn the significantly enhanced lysosomal escape and cytosol siRNA delivery. Therapeutic targeting of survivin was successfully applied for activation of apoptosis and programmed cell death. Combined photothermal and gene therapy improved therapeutic effectiveness.


Asunto(s)
Fosfatos de Calcio/química , Permeabilidad de la Membrana Celular , Hipertermia Inducida , Indoles/química , Nanopartículas/química , Fototerapia , Polímeros/química , ARN Interferente Pequeño/uso terapéutico , Animales , Calcificación Fisiológica , Muerte Celular , Terapia Combinada , Portadores de Fármacos/química , Endocitosis , Eritrocitos/metabolismo , Silenciador del Gen , Terapia Genética , Hemólisis , Células Hep G2 , Humanos , Lisosomas/metabolismo , Masculino , Ratones Desnudos , Nanopartículas/ultraestructura , Porosidad
13.
Mol Pharm ; 15(7): 2503-2512, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29768014

RESUMEN

Combination chemotherapy with a modulator and a chemotherapeutic drug has become one of the most promising strategies for the treatment of multidrug resistance (MDR) in cancer therapy. However, the development of nanocarriers with a high payload and sequential release of therapeutic agents poses a significant challenge. In this work, we report a type of hybrid nanocarriers prepared by polydopamine (PDA) mediated integration of the mesoporous MSN core and the microporous zeolite imidazolate frameworks-8 (ZIF-8) shell. The nanocarriers exploit storage capacities for drugs based on the high porosity and molecular sieving capabilities of ZIF-8 for sequential drug release. Particularly, large amounts of an anticancer drug (DOX, 607 µg mg-1) and a MDR inhibitor curcumin (CUR, 778 µg mg-1) were sequentially loaded in the mesoporous core via π-π stacking interactions mediated by PDA and in the microporous shell via the encapsulation during ZIF-8 growth. The sustained release of DOX was observed to follow earlier and faster release of CUR by acid-sensitive dissolution of the ZIF-8 shell. Furthermore, the nanoparticles showed good biocompatibility and effective cellular uptake in in vitro evaluations using drug-resistant MCF-7/ADR cancer cells. More importantly, the preferentially released CUR inhibited the drug efflux function of the membrane P-glycoprotein (P-gp), which subsequently facilitated the nuclear transportation of DOX released from the PDA-MSN core, and, in turn, the synergistic effects on killing MDR cancer cells. The hybrid mesoporous-microporous nanocarrier holds great promise for combination chemotherapy applications on the basis of sequential drug release.


Asunto(s)
Antineoplásicos/farmacología , Portadores de Fármacos/química , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Curcumina/farmacología , Curcumina/uso terapéutico , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Combinación de Medicamentos , Composición de Medicamentos/métodos , Liberación de Fármacos , Humanos , Células MCF-7 , Nanopartículas/química , Neoplasias/patología , Porosidad
14.
J Colloid Interface Sci ; 527: 68-77, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-29777974

RESUMEN

The photoluminescence stability of lanthanide complex in aqueous media is a prerequisite for diagnostics probes. The combination of building blocks working in concert to facilitate host-guest structures is now considered state of the art in surpassing this roadblock, yet there still remains a tremendous challenge. Here, a stable, highly-luminescent system was developed through trapping anionic complexes sensitized by tridentate pyridine-tetrazolate (pytz) ligands within the rigid framework of ZIF-8 (zeolitic imidazolate framework-8) particles (∼60 nm in size). The key to maintaining the stable luminescence of lanthanide complexes inside ZIF-8 frameworks is a stopcock design, i.e. stopper molecules (an imidazolium based ionic liquid) selectively plugged on the pore entrances located at the exterior surface of the ZIF-8 host, which protect both the host and the guests from deteriorations by surrounding ions/water molecules. Remarkably, the obtained Ln complex encapsulated ZIF-8 particles (Ln = terbium, europium) particles possessed high quantum yields (23.2% and 8.5%), large absorption cross-section (∼10-12 cm2), and long luminescence lifetimes (1.9 and 3.0 ms) in PBS buffer. In addition, the system can realize single/multi-color encoding by altering the loading amounts and the weight ratios of complexes emitting at different wavelengths.

15.
J Mater Chem B ; 6(12): 1754-1763, 2018 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-32254247

RESUMEN

Nanoscale colloidal capsules are promising drug delivery carriers currently while the demand for multiple-step syntheses and the difficulties in achieving high capsule stability are key obstacles that have greatly restricted their development. Herein, we report a polydopamine (PDA) nanoparticle stabilized nanocapsule as a drug delivery system based on the combination of nanoparticle formation and capsule assembly/stabilization in one pot. In this system, an arginine modified linoleic acid nanoemulsion was employed as the template for the in situ generation/assembly of interfacially active PDA nanoparticles, while directional interaction pairs of carboxylate-guanidine and amino-PDA linked by arginine are involved in the assembly process. The nanocapsules possess an average size of 100 nm, high stability in biological media, and efficient lipophilic transfer of the loaded lipophilic cargo. Notably, the high biocompatibility of the nanocapsules and the non-endocytotic delivery to the cytosol of cancer cells were demonstrated in vitro. Furthermore, the efficient delivery of paclitaxel, as well as paclitaxel/doxorubicin dual cargo, was realized, resulting in the high inhibition of cancer cells. Altogether, the PDA nanoparticle stabilized nanocapsules open new opportunities for the development of promising nanocapsule platforms for biomedical delivery.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...