Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(52): 32962-32969, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33318219

RESUMEN

Clinical investigations have shown that a nonimmunogenic "cold" tumor is usually accompanied by few immunopositive cells and more immunosuppressive cells in the tumor microenvironment (TME), which is still the bottleneck of immune activation. Here, a fluorine assembly nanocluster was explored to break the shackles of immunosuppression, reawaken the immune system, and turn the cold tumor "hot." Once under laser irradiation, FS@PMPt produces sufficient reactive oxygen species (ROS) to fracture the ROS-sensitive linker, thus releasing the cisplatin conjugated PMPt to penetrate into the tumors and kill the regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). Meanwhile, ROS will induce potent immunogenic cell death (ICD) and further promote the accumulation of dendritic cells (DCs) and T cells, therefore not only increasing the infiltration of immunopositive cells from the outside but also reducing the immunosuppressive cells from the inside to break through the bottleneck of immune activation. The FS@PMPt nanocluster regulates the immune process in TME from negative to positive, from shallow to deep, to turn the cold tumor into a hot tumor and provoke a robust antitumor immune response.


Asunto(s)
Antineoplásicos/síntesis química , Flúor/química , Factores Inmunológicos/síntesis química , Nanoconjugados/química , Microambiente Tumoral/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Dendrímeros/química , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Factores Inmunológicos/farmacología , Ratones , Ratones Endogámicos BALB C , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Platino (Metal)/química , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología
2.
Pharmacol Res ; 185: 106490, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36216131

RESUMEN

Acquired drug resistance and epithelial-mesenchymal transition (EMT) mediated metastasis are two highly interacting determinants for non-small-cell lung cancer (NSCLC) prognosis. This study investigated the common mechanisms of drug resistance and EMT from the perspective of metabolic reprogramming, which may offer new ideas to improve anticancer therapy. Acquired resistant cells were found to grow faster and have a greater migratory and invasive capacity than their parent cells. Metabolomics analysis revealed that acquired resistant cells highly relied on glutamine utilization and mainly fluxed into oxidative phosphorylation energy production. Further mechanistic studies screened out glutamate dehydrogenase 1 (GLUD1) as the determinant of glutamine addiction in acquired resistant NSCLC cells, and provided evidence that GLUD1-mediated α-KG production and the accompanying reactive oxygen species (ROS) accumulation primarily triggered migration and invasion by inducing Snail. Pharmacological and genetic interference with GLUD1 in vitro significantly reversed drug resistance and decreased cell migration and invasion capability. Lastly, the successful application of R162, a selective GLUD1 inhibitor, to overcome both acquired resistance and EMT-induced metastasis in vivo, identified GLUD1 as a promising and druggable therapeutic target for malignant progression of NSCLC. Collectively, our study offers a potential strategy for NSCLC therapy, especially for drug-resistant patients with highly expressed GLUD1.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal , Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/uso terapéutico , Glutamina/metabolismo , Glutamina/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Factores de Transcripción de la Familia Snail/metabolismo
3.
Angew Chem Int Ed Engl ; 61(22): e202202843, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35238124

RESUMEN

The abundant glutathione (GSH) in "cold" tumors weakens ferroptosis therapy and the immune response. Inspired by lipids, we fabricated cinnamaldehyde dimers (CDC) into lipid-like materials to form dimersomes capable of depleting GSH and delivering therapeutics to potentiate the ferroptosis and immunotherapy of breast cancer. The dimersomes exhibited superior storage stability for over one year. After reaching the tumor, they quickly underwent breakage in the cytosol owing to the conjugation of hydrophilic GSH on CDC by Michael addition, which not only triggered the drug release and fluorescence switch "ON", but also led to the depletion of intracellular GSH. Ferroptosis was significantly enhanced after combination with sorafenib (SRF) and elicited a robust immune response in vivo by promoting the maturation of dendritic cells and the priming of CD8+ T cells. As a result, the CDC@SRF dimersomes cured breast cancer in all the mice after four doses of administration.


Asunto(s)
Ferroptosis , Neoplasias , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Glutatión , Inmunoterapia , Ratones , Neoplasias/tratamiento farmacológico , Sorafenib/farmacología , Sorafenib/uso terapéutico
4.
Mol Pharm ; 18(2): 667-678, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32579365

RESUMEN

Gasdermin D (GSDMD) plays a causal role in NOD-like receptor protein 3 (NLRP3) inflammasome-mediated pyroptosis eruption, which has been regarded as a potential therapeutic target for pyroptosis-related diseases including acute gouty arthritis. In the present study, the synthesized PEI-Chol (cholesterol grafted polyethylenimine) was assembled with GSDMD small interfering RNA (siRNA) to form PEI-Chol/siGSDMD polyplexes, which provided high transfection efficiency for siRNA-mediated GSDMD knockdown. Then we evaluated the effect of GSDMD siRNA-loaded PEI-Chol on inflammatory cascades in bone-marrow-derived macrophages (BMDMs) and acute gouty arthritis animal models under MSU exposure. When accompanied by pyroptosis blockade and decreased release of interleukin-1 beta (IL-1ß), NLRP3 inflammasome activation was also suppressed by GSDMD knockdown in vivo and in vitro. Moreover, in MSU-induced acute gouty arthritis mice, blocking GSDMD with siRNA significantly improved ankle swelling and inflammatory infiltration observed in histopathological analysis. Furthermore, investigation using a mouse air pouch model verified the effect of siGSDMD-loaded PEI-Chol on pyroptosis of recruited macrophages and related signaling pathways in response to MSU. These novel findings exhibited that GSDMD knockdown relieved acute gouty arthritis through inhibiting pyroptosis, providing a possible therapeutic approach for MSU-induced acute gouty arthritis molecular therapy using PEI-Chol as a nucleic acid delivery carrier.


Asunto(s)
Artritis Gotosa/tratamiento farmacológico , Portadores de Fármacos/química , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas de Unión a Fosfato/antagonistas & inhibidores , Piroptosis/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Gotosa/inducido químicamente , Artritis Gotosa/inmunología , Artritis Gotosa/patología , Células Cultivadas , Colesterol , Técnicas de Silenciamiento del Gen/métodos , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Polietileneimina/química , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Ácido Úrico/administración & dosificación , Ácido Úrico/toxicidad
5.
Drug Dev Ind Pharm ; 47(3): 429-439, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33617404

RESUMEN

As a natural flavonoid compound, rutin could scavenge free radicals effectively to achieve remarkable antioxidant and anti-photoaging activity. Unfortunately, the extremely low water solubility of rutin often leads to the poor percutaneous permeability and unsatisfactory bioavailability, which has greatly restricted its clinical application. In this study, a novel freeze-dried rutin nanocrystal was developed to improve its saturation solubility, which was further redispersed in carbopol gel to formulate the targeted rutin nanocrystal gel (NC-gel) for enhanced transdermal delivery efficiency. Benefit from the advantages of NC-gel, the permeated amounts of rutin on mice in the NC-gel group was more than three times enhancement over that of the coarse drug gel group. Furthermore, the results of pharmacodynamic studies in vivo demonstrated that NC-gel could effectively prevent the skin photoaging and tissue damage induced by UV irradiation. Taken together, these results validated that NC-gel was an ideal carrier for the epidermal application of rutin to obtain excellent anti-photoaging effect, which further might provide a valuable platform for improving the transdermal bioavailability of insoluble drugs.


Asunto(s)
Nanopartículas , Rutina , Administración Cutánea , Animales , Disponibilidad Biológica , Ratones , Piel/metabolismo , Solubilidad
6.
Nano Lett ; 20(7): 4842-4849, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32578994

RESUMEN

Development of T1/T2 dual-mode MRI contrast agents that can also treat cancer is an attractive prospect for personalized precision medicine. Unfortunately, conventional contrast agents can suffer from toxicity and lack any ability to treat cancer. An all-iron T1/T2 MR imaging agent with photothermal and drug delivery capability would overcome these issues. Here, an avocado-like Fe3+/Fe2O3 composed T1-T2 dual-mode contrast agent based on Fe-TA coordination network (CNMN) is developed. This material possesses suitable longitudinal and transverse relaxation coefficients. Moreover, the strong heat generation property of Fe-TA endows CNMN with the capability to act as a potent photothermal agent. Furthermore, CNMN can also act as an effective delivery platform for the chemotherapeutic drug doxorubicin (DOX) to achieve high effective chemo-photothermal combination therapy. The work demonstrates reliable T1-T2 MRI-guided chemo-photothermal therapy for safe and effective clinical application.


Asunto(s)
Nanopartículas , Neoplasias , Persea , Doxorrubicina/uso terapéutico , Hierro , Imagen por Resonancia Magnética , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Fototerapia , Medicina de Precisión
7.
Bioconjug Chem ; 31(3): 698-707, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-31967460

RESUMEN

Polyethylenimine (PEI) is a promising delivery vector of nucleic acids, but cytotoxicity and only moderate transfection efficacy with small RNAs limit its applications. Here we hypothesized that hydrophobization of PEI by combined modification with perfluorinated moieties (F) and cholesterol (Ch) will help in addressing both the cytotoxicity and siRNA delivery efficacy. To test the hypothesis, we synthesized a series of copolymers (F-PEI-Ch) by modifying PEI by reaction with heptafluorobutyric anhydride and cholesteryl chloroformate. We investigated and compared the effect of the modifications on siRNA delivery in vitro and in vivo. We found that the F-PEI-Ch copolymers assembled into micellar structures and that the copolymer with the highest Ch content exhibited the best siRNA delivery performance, including lower cytotoxicity, enhanced cell uptake, improved endosomal escape, and the best siRNA silencing efficacy in vitro and in vivo when compared with control PEI, F-PEI, and PEI-Ch. Overall, hydrophobization of PEI with a combination of cholesterol and superhydrophobic perfluorinated moieties represents a promising approach to the design of siRNA delivery vectors with decreased toxicity and enhanced transfection efficacy.


Asunto(s)
Colesterol/química , Portadores de Fármacos/química , Fluorocarburos/química , Interacciones Hidrofóbicas e Hidrofílicas , Polietileneimina/química , ARN Interferente Pequeño/química , Animales , Línea Celular Tumoral , Silenciador del Gen , Ratones , ARN Interferente Pequeño/genética
8.
Mol Ther ; 27(12): 2100-2110, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31481310

RESUMEN

Lung metastasis is a common and deadly occurrence in many types of solid tumors. Chemokine receptor CXCR4 and transcription factor signal transducer and activator of transcription 3 (STAT3) are among potential therapeutic targets in lung metastatic cancer. Both CXCR4 and STAT3 play important roles in the proliferation, angiogenesis, and metastasis of cancer cells. Here, we report on the development of a pulmonary delivery (p.d.) system based on perfluorocarbon (PFC) nanoemulsions for combined delivery of a partially fluorinated polymeric CXCR4 antagonist (FM) and anti-STAT3 small interfering RNA (siRNA). We have prepared FM-stabilized PFC (FM@PFC) as a delivery system of therapeutic siRNA adsorbed on the surface of the emulsion. These FM@PFC/siRNA nanoemulsions inhibited both CXCR4 and STAT3, as demonstrated by effective anti-invasive ability in vitro and related antimetastatic activity in vivo. The combined nanoemulsions provided a comprehensive anticancer effect in the model of established lung metastasis of breast carcinoma, which was dependent on induction of cancer cell apoptosis, anti-angiogenic effect, anti-invasive activity, and overcoming of the immunosuppressive tumor microenvironment. Direct comparison with intravenous (i.v.) injection showed superior activity of pulmonary administration as indicated by significantly increased animal survival. Overall, this work established the suitability of the PFC nanoemulsions for p.d. of combination anticancer treatments and as a promising method to treat lung metastasis.


Asunto(s)
Neoplasias de la Mama/terapia , Sistemas de Liberación de Medicamentos , Neoplasias Pulmonares/terapia , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Receptores CXCR4/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Nanopartículas/química , Neovascularización Patológica , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/genética , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Nanomedicine ; 23: 102071, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31442581

RESUMEN

Developing various kinds of nanoplatforms with integrated diagnostic and therapeutic functions would be significant for imaging-guided precision treatment of cancer. However, it is still a challenge to organically integrate therapeutic and imaging components into a single nano-system rather than simply mixing. Herein, an iron-gallic acid network-based nanoparticle (Fe-GA@PEG-PLGA) was designed for magnetic resonance imaging (MRI)-guided chemo-photothermal synergistic therapy of tumors. The tumor spatial location and size information can be accurately achieved due to T1 MRI based on Fe3+ coordination with GA in Fe-GA network. Furthermore, the nanoparticle exhibited extraordinary photostability and photothermal therapy capacity exceeded 42 °C within 100 s under 808 nm laser irradiation. Meanwhile, the Fe-GA polymeric network can be disassembled in tumor acidic environment and the released drug GA can induce apoptosis. This study demonstrated that the Fe-GA network-based nanoparticle is a promising diagnostic and therapeutic agent for theranostic application and further clinic translation.


Asunto(s)
Imagen por Resonancia Magnética , Nanoestructuras , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/terapia , Fototerapia , Animales , Ácido Gálico/química , Ácido Gálico/farmacología , Humanos , Concentración de Iones de Hidrógeno , Hipertermia Inducida , Hierro/química , Hierro/farmacología , Ratones , Nanoestructuras/química , Nanoestructuras/uso terapéutico
10.
Nano Lett ; 19(7): 4334-4342, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31179709

RESUMEN

Nanozymes as artificial enzymes that mimicked natural enzyme-like activities have received great attention in cancer diagnosis and therapy. Biomimetic nanozymes require more consideration regarding complicated tumor microenvironments to mimic biological enzymes, thus achieving superior nanozyme activity in vivo. Here we report a biomimetic hybrid nanozyme (named rMGB) which integrates natural enzyme glucose oxidase (GOx) with nanozyme manganese dioxide (MnO2) by mutual promotion for maximizing the enzymatic activity of MnO2 and GOx. Under hypoxia environment, we observed that MnO2 could react with endogenous H2O2 to produce O2 for enhancing the catalytic efficiency of GOx for starvation therapy. Meanwhile, we confirmed that glucose oxidation generated gluconic acid and further improved the catalytic efficiency of MnO2 subsequently. The biochemical reaction cycle, consisting of MnO2, O2, GOx, and H+, was triggered by the tumor microenvironment and accelerated each other so as to achieve self-supplied H+ and accelerate O2 generation, enhancing the starvation therapy, alleviating tumor hypoxia and accelerating the reactive oxygen species generation in photodynamic therapy. This biomimetic hybrid nanozyme would further facilitate the development of biological nanozymes for cancer treatment.


Asunto(s)
Materiales Biomiméticos , Glucosa Oxidasa , Compuestos de Manganeso , Nanoestructuras , Neoplasias Experimentales , Óxidos , Oxígeno/metabolismo , Fotoquimioterapia , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Glucosa Oxidasa/química , Glucosa Oxidasa/farmacología , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Ratones , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Óxidos/química , Óxidos/farmacología , Especies Reactivas de Oxígeno/metabolismo
11.
Mol Pharm ; 16(6): 2728-2741, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31070930

RESUMEN

Peritoneal fibrosis (PF) is a common complication of long-term peritoneal dialysis (PD). It is considered as the main reason for dialysis inadequacy and PD withdrawal. Transforming growth factor beta (TGF-ß) regulates the expression of stromal cell-derived factor 1 (SDF-1α) and its receptor C-X-C chemokine receptor type 4 (CXCR4) on human peritoneal mesothelial cells (HPMCs), resulting in an increased migratory potential of HPMCs and extracellular matrix (ECM) deposition in the scar tissue and eventually fibrosis. Because SDF-1α/CXCR4 activation has a vital role in the pathogenesis of PF, codelivery of a CXCR4-receptor targeting agent with an antifibrotic agent in a single nanocarrier can be a promising strategy for treating PF. Here, for the first time, AMD3100 (AMD), a CXCR4-receptor antagonist, was coformulated with sulfotanshinone IIA sodium (STS IIA) into a liposome (STS-AMD-Lips) to develop a CXCR4 receptor targeting form of combination therapy for PF. CXCR4 targeting increased the ability of liposomes to target fibrotic peritoneal mesothelial cells overexpressing CXCR4 and facilitated the ability of STS IIA treatment at the fibrotic site. The liposome had an average diameter of 103 nm with encapsulated efficiencies of above 50%. The in vivo studies confirmed the reversal of PD solution-induced epithelial-to-mesenchymal transition by STS-AMD-Lips in HPMCs. The in vivo studies also revealed the precise biodistribution of the liposomes to peritoneum. Significant reduction of the morphological lesions and decreased level of ECM proteins were observed in rats treated with STS-AMD-Lips, proving that the liposomal nanocarrier has excellent ability to reverse PF. It has been concluded that the STS-AMD-Lips exhibit specific peritoneal targeting ability and could be used to improve STS-AMD combination delivery for the treatment of PF.


Asunto(s)
Liposomas/uso terapéutico , Fibrosis Peritoneal/tratamiento farmacológico , Fibrosis Peritoneal/metabolismo , Receptores CXCR4/metabolismo , Animales , Western Blotting , Supervivencia Celular/fisiología , Quimiocina CXCL12/metabolismo , Femenino , Citometría de Flujo , Humanos , Inmunoquímica , Ratones Endogámicos BALB C
12.
Cell Physiol Biochem ; 46(6): 2434-2444, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29742502

RESUMEN

BACKGROUND/AIMS: Long-term use of high-glucose peritoneal dialysis solution (PDS) induces peritoneal mesothelial cell (PMC) injury, peritoneal dysfunction, and peritoneal dialysis (PD) failure in patients with end-stage renal disease. How to preserve PMCs in PD is a major challenge for nephrologists worldwide. In this study, we aimed to elucidate the efficacy and mechanisms of sulfotanshinone IIA sodium (Tan IIa) in ameliorating high-glucose PDS-induced human PMC injury. METHODS: The human PMC line HMrSV5 was incubated with 4.25% PDS in vitro to mimic the high-glucose conditions in PD. Cellular viability was measured by Cell Counting Kit 8. Generation of superoxide and reactive oxygen species (ROS) was assessed using a Total ROS/Superoxide Detection Kit. Oxidative modification of protein was evaluated by OxyBlot Protein Oxidation Detection Kit. TUNEL (dT-mediated dUTP nick end labeling) assay and DAPI (4,6-diamidino-2-phenylindole) staining were used to evaluate apoptosis. Western blot analysis was performed to evaluate the efficacy and mechanisms of Tan IIa. RESULTS: Tan IIa protected PMCs against PDS-induced injury as evidenced by alleviating changes in morphology and loss of cell viability. Consistent with their antioxidant properties, N-acetyl-L-cysteine (NAC) and Tan IIa suppressed superoxide and ROS production, protein oxidation, and apoptosis elicited by PDS. Apoptosis signal-regulating kinase 1 (ASK1)-p38 signaling was activated by PDS. Both Tan IIa and NAC suppressed ASK1 and p38 phosphorylation elicited by PDS. Moreover, genetic downregulation of ASK1 ameliorated cell injury and inhibited the phosphorylation of p38 and activation of caspase 3. CONCLUSION: Tan IIa protects PMCs against PDS-induced oxidative injury through suppression of ASK1-p38 signaling.


Asunto(s)
Soluciones para Diálisis/efectos adversos , Medicamentos Herbarios Chinos/farmacología , Glucosa/efectos adversos , Diálisis Peritoneal/efectos adversos , Peritoneo/efectos de los fármacos , Sustancias Protectoras/farmacología , Salvia miltiorrhiza/química , Transducción de Señal/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Humanos , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Diálisis Peritoneal/métodos , Peritoneo/citología , Peritoneo/metabolismo , Peritoneo/patología
13.
Biomacromolecules ; 19(9): 3776-3787, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30081638

RESUMEN

Cationic polyplex as commonly used nucleic acid carriers faced several shortcomings, such as high cytotoxicity, low serum stability, and slow cargo release at the target site. The traditional solution is covering a negative charged layer (e.g., hyaluronic acid, HA) via electrostatic interaction. However, it was far from satisfactory for the deshielding by physiological anions in circulation (e.g., serum proteins, phosphate). In this study, we proposed a new strategy of reversible covalent cross-linking to enhance stability in circulation and enable stimuli-disassembly of polyplexes in tumor cells. Here, 25k polyethylenimine (PEI) was chosen as model polycations for veriying the hypothesis. HA-PEI conjugation was formed by the cross-linking of adenosine triphosphate grafted HA (HA-ATP) with phenylboronic acid grafted PEI (PEI-PBA) via the chemical reaction between PBA and ATP. Compared with noncovalent polyplex by electrostatic interaction (HA/PEI), HA-PEI exhibited much better colloidal stability and serum stability. The covered HA-ATP layer on PEI-PBA could maintain stable in the absence of physiological anions, while HA layer on PEI in HA/PEI group showed obvious detachment after anion's competition. More importantly, the covalent cross-linking polyplex could selectively release siRNA in the ATP rich environment of cytosol and significantly improve siRNA silence. Besides, the covalent cross-linking with HA-ATP could effectively reduce the cytotoxicity of cationic polyplex, improve the uptake by B61F10 cells and promote the endosomal escape. Consequently, this strategy of HA-PEI conjugation significantly enhanced the siRNA transfection in the absence or presence of FBS (fetal bovine serum) on B16F10 cells and CHO cells. Taken together, the reversible covalent cross-linking approach shows obvious superiority compared with the noncovalent absorption strategy. It held great potential to be developed to polish up the performance of cationic polyplex on reducing the toxicity, enhancing the serum tolerance and achieving controlled release of siRNA at target site.


Asunto(s)
Adenosina Trifosfato/química , Reactivos de Enlaces Cruzados/química , Técnicas de Transferencia de Gen , Poliaminas/química , Interferencia de ARN , Animales , Ácidos Borónicos/química , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Ácido Hialurónico/química , Masculino , Ratones , Ratones Endogámicos C57BL , Polielectrolitos , Polietileneimina/química , Tratamiento con ARN de Interferencia/métodos
14.
Biomacromolecules ; 19(2): 392-401, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29350899

RESUMEN

Chemokine receptor CXCR4 plays an important role in cancer cell invasion and metastasis. Recent findings suggest that anti-VEGF therapies upregulate CXCR4 expression, which contributes to resistance to antiangiogenic therapies. Here, we report the development of novel derivatives of polyethylenimine (PEI) that effectively inhibit CXCR4 while delivering anti-VEGF siRNA. PEI was alkylated with different amounts of a CXCR4-binding cyclam derivative to prepare PEI-C. Modification with the cyclam derivatives resulted in a considerable decrease in cytotoxicity when compared with unmodified PEI. All the PEI-C showed significant CXCR4 antagonism and the ability to inhibit cancer cell invasion. Polyplexes of PEI-C prepared with siVEGF showed effective silencing of the VEGF expression in vitro. In vivo testing in a syngeneic breast cancer model showed promising antitumor and antimetastatic activity of the PEI-C/siVEGF polyplexes. Our data demonstrate the feasibility of using PEI-C as a carrier for simultaneous VEGF silencing and CXCR4 inhibition for enhanced antiangiogenic cancer therapies.


Asunto(s)
Antineoplásicos/química , Compuestos Heterocíclicos/química , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Polietileneimina/química , Tratamiento con ARN de Interferencia/métodos , Receptores CXCR4/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Femenino , Silenciador del Gen , Humanos , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Nanomedicine ; 14(6): 1765-1776, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29777878

RESUMEN

This report describes the development of polyplexes based on CXCR4-inhibiting poly(ethylenimine) derivative (PEI-C) for pulmonary delivery of siRNA to silence plasminogen activator inhibitor-1 (siPAI-1) as a new combination treatment of pulmonary fibrosis (PF). Safety and delivery efficacy of the PEI-C/siPAI-1 polyplexes was investigated in vitro in primary lung fibroblasts isolated from mice with bleomycin-induced PF. Biodistribution analysis following intratracheal administration of fluorescently labeled polyplexes showed prolonged retention in the lungs. Treatment of mice with bleomycin-induced PF using the PEI-C/siPAI-1 polyplexes resulted in a significant down-regulation of the PAI-1 expression and decreased collagen deposition in the lung. The results of this study provide first evidence of the potential benefits of combined inhibition of CXCR4 and PAI-1 in the pulmonary treatment of PF.


Asunto(s)
Sistemas de Liberación de Medicamentos , Polietileneimina/química , Polímeros/administración & dosificación , Fibrosis Pulmonar/prevención & control , ARN Interferente Pequeño/genética , Receptores CXCR4/antagonistas & inhibidores , Serpina E2/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/toxicidad , Apoptosis , Bleomicina/toxicidad , Proliferación Celular , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Silenciador del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Polímeros/química , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Receptores CXCR4/genética , Serpina E2/genética
16.
Nanomedicine ; 14(7): 2283-2294, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29981460

RESUMEN

Tumor angiogenesis is a key step in the process of tumor development, and antitumor angiogenesis has a profound influence on tumor growth. Herein we report a dual-function drug delivery system comprising a Near-infrared (NIR) dye and an anti-angiogenic drug within liposomes (Lip-IR780-Sunitinib) for enhanced antitumor therapy. The hydrophobic NIR dye IR780 was loaded into the liposome phospholipid bilayer, and the bilayer would be disrupted by laser irradiation so that anti-angiogenic drug sunitinib release would be activated remotely at the tumor site. The released hydrophilic sunitinib could potentially target multiple VEGF receptors on the tumor endothelial cell surface to inhibit angiogenesis. Meanwhile, IR780-loaded liposomes kill the cancer cells by photothermal therapy. Lip-IR780-Sunitinib exhibited enhanced anti-tumor and anti-angiogenic effects in vitro and in vivo. This system facilitates easy and controlled release of cargos to achieve anti-tumor angiogenesis and photothermal therapy.


Asunto(s)
Neoplasias de la Mama/terapia , Hipertermia Inducida , Indoles/química , Liposomas/administración & dosificación , Neovascularización Patológica/terapia , Fototerapia , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Terapia Combinada , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Indoles/administración & dosificación , Rayos Láser , Liposomas/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/patología , Espectroscopía Infrarroja Corta , Sunitinib/química , Sunitinib/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Biomacromolecules ; 18(6): 1793-1802, 2017 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-28445650

RESUMEN

The unsatisfied results of cancer therapy are caused by many issues and metastasis of cancer cells is one of the major challenge. It has been reported that inhibiting the SDF1/CXCR4 interaction can significantly reduce the metastasis of breast cancer cells to regional lymph nodes and lung. Herein, a nanogel system equipped with the FDA-approved CXCR4 antagonist AMD3100 was developed and evaluated for its combined antimetastatic and tumor targeting effects. Briefly, a bioreducible cross-linked dextrin nanogel (DNG) coated with AMD3100 was designed to possess multiple functions, including CXCR4 chemokine targeting, inhibition of tumor metastasis, and reduction-responsive intracellular release of doxorubicin (DOX) to reduce the cells proliferation. The in vitro results confirmed that the DOX-loaded AMD3100-coated dextrin nanogel (DOX-AMD-DNG) was more effectively taken up by 4T1 breast cancer cells than DOX-DNG and was significantly more cytotoxic to 4T1 cells than DOX-DNG. In biodistribution studies, the stronger fluorescence intensity of Cy7-AMD-DNG than Cy7-DNG further confirmed that AMD3100 mediated tumor targeting in vivo. AMD3100-coated DOX-DNG also exhibited a distinct antimetastatic effect and CXCR4 antagonistic activity by inhibiting CXCR4-mediated cell invasion in 4T1 and U2OS cells. Moreover, DOX-AMD-DNG displayed superior anticancer activity and antimetastatic effects in orthotopic breast cancer-bearing Balb/C mice. In summary, the multifunctional DOX-AMD-DNG can effectively target the tumor site and dually impede cancer progression and metastasis.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/farmacología , Portadores de Fármacos , Compuestos Heterocíclicos/farmacología , Nanoestructuras/química , Receptores CXCR4/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/química , Bencilaminas , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Ciclamas , Dextrinas/química , Doxorrubicina/química , Femenino , Geles , Expresión Génica , Compuestos Heterocíclicos/química , Humanos , Ratones , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Nanoestructuras/administración & dosificación , Nanoestructuras/ultraestructura , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Oxidación-Reducción , Unión Proteica , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Plant Foods Hum Nutr ; 69(3): 270-5, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24913513

RESUMEN

This study assessed the effects of thermal (40, 60, 80, 100 and 127 °C) and high hydrostatic pressure (HHP, 200, 400 and 600 MPa) treatments on the in vitro digestibility and structural properties of sweet potato protein (SPP). The results showed that the in vitro digestibility of SPP increased significantly with increasing heating temperature and heating time (0-60 min), while HHP treatment had little or no effect. Native SPP denaturation temperature (T d ) and enthalpy change (ΔH) were 89.0 °C and 9.6 J/g, respectively. Thermal and HHP treated SPP had T d of 84.6-88.9 °C and 86.4-87.6 °C, respectively. ΔH of thermal treated SPP was 3.6-6.4 J/g, while that of HHP treated SPP was 5.9-7.8 J/g. The differential scanning calorimetry (DSC) results demonstrated that HHP and thermal treatments both significantly reduced SPP thermodynamic stability. Circular dichroism analyses revealed that native SPP contains α-helixes, ß-sheets and random coils (4.3, 48.0 and 47.7%, respectively). After thermal treatment at 127 °C for 20 min, the content of α-helixes and turns increased significantly (13.2 and 27.6%, respectively), whereas the content of ß-sheets decreased significantly (12.3%). In contrast, HHP treatment increased the content of ß-sheets, but decreased the content of random coils. This study suggested that the SPP structure changes might be the main reason affecting the in vitro digestibility of SPP, and thermal treatment was more effective at changing SPP secondary structures and improving in vitro SPP digestibility than HHP treatment.


Asunto(s)
Calor , Ipomoea batatas/química , Proteínas de Plantas/química , Rastreo Diferencial de Calorimetría , Dicroismo Circular , Digestión , Manipulación de Alimentos , Presión Hidrostática , Pliegue de Proteína , Estructura Secundaria de Proteína
19.
Mol Pharm ; 10(6): 2479-89, 2013 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-23646913

RESUMEN

Refractory leukemia remains the most common therapeutic problem in clinical treatment of leukemia. The key therapy of refractory leukemia is to kill, thoroughly, the minimal residual disease and leukemia stem cells in the highly vascularized red marrow areas. In this study, two new conjugates, alendronate-polyethylene glycol (100) monostearate and folate-polyethylene glycol (100) monostearate, were synthesized to develop a multistep targeting nanostructured lipid carriers by enhancing drug transport to the high bone turnover areas adjacent to the red marrow and targeting the minimal residual disease and leukemia stem cells. This dual targeting system demonstrated a great binding affinity to hydroxyapatite, a model component of bone minerals, and higher cell uptake (in the form of carriers but not drug) and cytotoxicity in the K562 cell line, a leukemia cell line with overexpressed folate receptors, were observed in vitro compared to unmodified carriers, especially when the cells were pretreated and the receptors were up-regulated by all-trans retinoic acid. The comodel test of K562 cells and HA showed that this dual targeting system could desorb from bone surface and be taken up by leukemia cells. For the in vivo study, this dual targeting system exhibited a significant increase in plasma half-life and could specifically accumulate in the bone tissue of rats or mice after intravenous injection. Ex vivo imaging of mice femurs and confocal laser scanning microscope imaging of mice femur slices further confirmed that this dual targeting system could favorably deposit to the osteoblast-enriched areas of high bone turnover in regions of trabecular bone surrounded by red marrow. In vivo antitumor activity in K562/BALB/c-nu leukemia mice showed that the treatment of this dual targeting system significantly reduced the white blood cell (WBC) number in peripheral blood and bone marrow to the normal level. In conclusion, this dual targeting system could precisely target to the regions where the minimal residual disease and leukemia stem cells are located and then be specifically uptaken in large amounts, which is a valuable target for refractory leukemia therapy.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Leucemia/tratamiento farmacológico , Neoplasia Residual/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Médula Ósea/efectos de los fármacos , Línea Celular Tumoral , Durapatita/química , Ácido Fólico/análogos & derivados , Ácido Fólico/química , Humanos , Leucocitos/efectos de los fármacos , Ratones , Mitoxantrona/administración & dosificación , Mitoxantrona/uso terapéutico , Polietilenglicoles/química , Ratas
20.
Medicine (Baltimore) ; 102(20): e33779, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37335711

RESUMEN

BACKGROUND: Gastroesophageal reflux-related chronic cough (GERC), is one common type of chronic cough. Drug treatment is effective for some GERC patients. But, there is refractory GERC (rGERC). For rGERC, fundoplication may be the only effective method. However, there were very few studies about laparoscopic fundoplication in treating rGERC, and the cure rate of fundoplication in treating rGERC was unknown. So there is a question, what is the cure rate of fundoplication in treating rGERC? To solve this question, we performed this meta-analysis. METHODS: The PRISMA strategy and Cochrane collaboration method were used for this study. Our study was registered with PROSPERO (ID: CRD42021251072). We searched PubMed, Medline, Web of Science, and the Cochrane databases from 1990 to December 2022. The meta-analysis was performed with Review Manager 5.4 and Stata 14. RESULTS: After selection and exclusion, 8 articles out of 672 were included. The meta-analysis showed the cure rate of laparoscopic fundoplication in treating rGERC was 62% (95% confidence interval: 53-71%), with no deaths in 503 patients. There was no significant heterogeneity or bias in the meta-analysis. CONCLUSIONS: In terms of safety, laparoscopic fundoplication is quite reliable offered by skilled surgeons. In terms of cure rate, laparoscopic fundoplication could completely heal two-thirds of rGERC patients; however, there are still some patients who can not be completely cured by fundoplication.


Asunto(s)
Reflujo Gastroesofágico , Laparoscopía , Humanos , Fundoplicación/métodos , Tos/etiología , Tos/cirugía , Laparoscopía/métodos , Reflujo Gastroesofágico/complicaciones , Reflujo Gastroesofágico/cirugía , Enfermedad Crónica , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA