Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Healthc Mater ; 13(15): e2400113, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38412500

RESUMO

Recently, nanotechnology-based drug delivery platforms in treating pulmonary arterial hypertension (PAH) have gradually emerged. However, large mechanical stress and shear stress in blood vessels greatly affect the retention of nanopreparative materials at lesion sites, severely limiting nanotechnology-based drug delivery. Herein, a stimuli-responsive nanocraft is rationally designed by actively anchoring E-selectin overexpressed on pulmonary arterial endothelial cells (PAECs), under hypoxic conditions, allowing effective accumulation and retention of the drug at the lesion site. Briefly, a nitrobenzene group is incorporated into the framework of a nanocarrier, and then it is simultaneously linked with chitosan. Additionally, the surface of the nanocarrier with sialic acid (SA) and encapsulated the clinically used drug ambrisentan (Am), which enables the anchoring of E-selectin and subsequent drug delivery is modifed. This system facilitates intercellular transport to pulmonary artery smooth muscle cells (PASMCs) when targeting PAECs and specifically responds to a reductive hypoxic microenvironment with elevated nitroreductase in PASMCs. Moreover, compared with free Am, nanoencapsulation and SA-PEG2000-NH2 prolong the blood circulation time, achieving better therapeutic outcomes in preventing vascular remodeling and reversing systolic dysfunction. The originality and contribution of this work reveal the promising value of this pulmonary arterial anchoring stimuli-responsive nanocraft as a novel therapeutic strategy for satisfactory PAH treatment.


Assuntos
Hipertensão Pulmonar , Miócitos de Músculo Liso , Artéria Pulmonar , Animais , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/citologia , Artéria Pulmonar/efeitos dos fármacos , Fenilpropionatos/química , Fenilpropionatos/farmacologia , Quitosana/química , Vasoconstrição/efeitos dos fármacos , Selectina E/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Nanopartículas/química , Hipóxia/metabolismo , Humanos , Masculino , Sistemas de Liberação de Medicamentos/métodos , Ratos , Ratos Sprague-Dawley , Camundongos , Piridazinas
2.
Nanoscale ; 15(27): 11625-11646, 2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37377137

RESUMO

Ischemic stroke is characterized by high morbidity, disability, and mortality. Unfortunately, the only FDA-approved pharmacological thrombolytic, alteplase, has a narrow therapeutic window of only 4.5 h. Other drugs like neuroprotective agents have not been clinically used because of their low efficacy. To improve the efficacy of neuroprotective agents and the effectiveness of rescue therapies for hyperacute ischemic stroke, we investigated and verified the variation trends of the blood-brain barrier (BBB) permeability and regional cerebral blood flow over 24 h in rats that had ischemic strokes. Hypoperfusion and the biphasic increase of BBB permeability are still the main limiting factors for lesion-specific drug distribution and drug brain penetration. Herein, the nitric oxide donor hydroxyurea (HYD) was reported to downregulate the expression of tight junction proteins and upregulate intracellular nitric oxide content in the brain microvascular endothelial cells subjected to oxygen-glucose deprivation, which was shown to facilitate the transport of liposomes across  brain endothelial monolayer in an in vitro model. HYD also increased the BBB permeability and promoted microcirculation in the hyperacute phase of stroke. The neutrophil-like cell-membrane-fusogenic hypoxia-sensitive liposomes exhibited excellent performance in targeting the inflamed brain microvascular endothelial cells, enhancing cell association, and promoting rapid hypoxic-responsive release in the hypoxic microenvironment. Overall, the combined HYD and hypoxia-sensitive liposome dosing regimen effectively decreased the cerebral infarction volume and relieved neurological dysfunction in rats that had ischemic strokes; these therapies were involved in the anti-oxidative stress effect and the neurotrophic effect mediated by macrophage migration inhibitory factor.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Fármacos Neuroprotetores , Acidente Vascular Cerebral , Ratos , Animais , Lipossomos/metabolismo , Hidroxiureia/farmacologia , Hidroxiureia/metabolismo , Hidroxiureia/uso terapêutico , AVC Isquêmico/metabolismo , Fármacos Neuroprotetores/farmacologia , Células Endoteliais , Encéfalo/metabolismo , Barreira Hematoencefálica/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo , Hipóxia , Isquemia Encefálica/tratamento farmacológico
3.
Mater Today Bio ; 18: 100532, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36691607

RESUMO

Ferroptosis as promising antitumor therapy strategy could be comprised by intracellular antioxidants, especially GSH and thioredoxin (Trx). They are both cofactors of Gpx4, the enzyme catalyzing the production of lipid peroxides to relieve oxidative stress, which drives the acquired ferroptosis resistance in tumors. Herein, the NADPH-consuming micelles are specially designed, which could collaborate with the ROS generating photodynamics therapy (PDT) by depleting intracellular GSH and Trx under hypoxia condition, resulting in ruined redox homeostasis and the final cascade amplified ferroptosis. The tailored micelle was briefly prepared by conjugating hypoxia-sensitive segment p-nitrobenzyl chloroformate (PNZ-Cl) to the hydrophilic chitosan (CS), the resulting micelle was further modified with photosensitizer Ce6 via PEG linkage. When receiving laser irradiation, the photosensitizer would generate ROS and consume oxygen in the meanwhile. The resulting anabatic hypoxia in turns promote the NTR-catalyzed electron-accepting response of micelles, with evidently enhanced NADPH consumption and ultimately ruined redox homeostasis, contributing to cascade amplified ferroptosis with robust ROS. Most importantly, the accompanied immunogenic cell death (ICD) and releasing danger-associated molecular patterns (DAMPs) could boost dendritic cells (DCs) maturation and the subsequent T-cell-mediated profound immune response. Collectively, the work excavates the other biochemical reaction during the hypoxia-sensitive process of C-N-Ce6 by diminishing intracellular GSH and Trx, providing a candidate of ferroptosis inducers against solid tumors.

4.
Curr Med Chem ; 30(12): 1368-1385, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35726412

RESUMO

Chitosan, the only naturally occurring polycationic polysaccharide derived from chitin, has long case been implicated in the designs of nanosystems for diverse biomedical and pharmaceutical applications owing to its exclusive biodegradability, biocompatibility, cationic property, and functional groups. Particularly, some intrinsic characteristics of chitosan equip it with high potential for facile preparation, flexible functionalization, and modification, which circumvent the defects of chitosan and account for extensive attempts in cancer therapy and theranostic. In this review, we first give a classifiable explanation of strategies in fabricating rationally-designed chitosan-based polymeric nanomaterials for cancer therapy, which are categorized by the physical, chemical, and biological intrinsic characteristics of chitosan, respectively. Specifically, examples harnessing the cationic charge of chitosan are clarified, and the accompanied pH-responsive ability functions frequently are also mentioned. Besides, strategies toward the modification of functional groups (amino and hydroxyl groups) in repeated glycosidic units of chitosan and their additional roles are also discussed here. Lastly, the biological superiority of chitosan as an adjuvant or a ligand for glycoprotein and the application of chitosan- based polymeric nanomaterials in theranostic are summarized. Altogether, this review provides a comprehensive overview of recent advances in chitosan-based polymeric nanomaterials for cancer therapy and theranostics from a brand new perspective.


Assuntos
Quitosana , Nanoestruturas , Neoplasias , Humanos , Quitosana/uso terapêutico , Quitosana/química , Medicina de Precisão , Nanoestruturas/uso terapêutico , Nanoestruturas/química , Sistemas de Liberação de Medicamentos , Polímeros/uso terapêutico , Neoplasias/tratamento farmacológico
5.
AAPS PharmSciTech ; 23(6): 167, 2022 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-35711068

RESUMO

Hypoxia is an important pathological phenomenon, and it can induce many tumor microenvironment changes, such as accumulations of intracellular lactic acid, decrease of tumor microenvironment pH value, and regulate a series of physiological and pathological processes such as adhesion, metastasis, and immune escape. Hypoxic tumor cells act as a key target for treating tumor. In this research, we designed and prepared PEG-nitroimidazole grafts, PEG-NI, and FA-PEG-NI. We first explored their physical and chemical properties to serve as a drug carrier. Then, the hypoxia-sensitive properties such as particle size changes and drug release were investigated. Finally, the tumor targeting ability was studied in vitro and in vivo, and anti-tumor capacity was determined. Both grafts showed excellent property as a nanodrug carrier and showed favorable drug encapsulation ability of sorafenib with the help of the hydrophobic chain of 6-(BOC-amino) hexyl bromide. The micelles responded to the hypoxic tumor environment with chemical and spatial structure changes leading to sensitive and fast drug release. With the modification of folic acid, FA-PEG-NI gained tumor targeting ability in vivo. FA-PEG-NI graft proved a potential targeting drug delivery system in the treatment of hypoxic hepatocellular carcinoma.


Assuntos
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Nitroimidazóis , Antineoplásicos/química , Carcinoma Hepatocelular/tratamento farmacológico , Linhagem Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Ácido Fólico/química , Humanos , Hipóxia/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Micelas , Polietilenoglicóis/química , Microambiente Tumoral
6.
J Zhejiang Univ Sci B ; 23(2): 171-172, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35187891

RESUMO

The online version of the original article can be found at https://doi.org/10.1631/jzus.B1900468 The original version of this article (Liu et al., 2020) unfortunately contained some mistakes. 1. Figs. 7c and 7d in p.229 were incorrect. The upper left and bottom left pictures in Fig. 7c were accidentally duplicated with the pictures at the same position of Fig. 1a. The upper right and bottom right pictures were mistakenly placed in Fig. 7c. Therefore, the calculation results in Fig. 7d were also mistaken. The correct versions should be as follows: 2. Because of the wrong pictures of Fig. 7c, the calculated results of "42.5%" in Abstract, Sections 3.9 and 5 are also mistaken. The correct result should be "45.2%." (1) Lines 10-12 of Abstract in p.218: "CSO-ss-SA/siRNA could effectively transmit siRNA into tumor cells, reducing the expression of RAC1 protein by 38.2% and decreasing the number of tumor-induced invasion cells by 42.5%." was incorrect. The correct version should be "CSO-ss-SA/siRNA could effectively transmit siRNA into tumor cells, reducing the expression of RAC1 protein by 38.2% and decreasing the number of tumor-induced invasion cells by 45.2%." (2) Lines 23-26 of Section 3.9 in p.227: "It was shown that the number of invasive tumor cells induced by DOX was reduced by 42.5% since CSO-ss-SA/siRNA downregulated the expression of RAC1 protein." was incorrect. The correct version should be "It was shown that the number of invasive tumor cells induced by DOX was reduced by 45.2% since CSO-ss-SA/siRNA downregulated the expression of RAC1 protein." (3) Lines 4-8 of Section 5 in p.231: "CSO-ss-SA, as an efficient redox-sensitive carrier for delivering siRNA silencing RAC1 into tumor cells, reduced the expression of RAC1 by 38.2% and decreased DOX-induced tumor invasion cells by 42.5% in vitro." was incorrect. The correct version should be "CSO-ss-SA, as an efficient redox-sensitive carrier for delivering siRNA silencing RAC1 into tumor cells, reduced the expression of RAC1 by 38.2% and decreased DOX-induced tumor invasion cells by 45.2% in vitro."

7.
Biomaterials ; 275: 120958, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34130142

RESUMO

Tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) play an important role in the development of tumors by secreting a variety of cytokines or directly communicating with tumor cells, making TAMs-targeted therapeutic strategies very attractive. It has been reported that oncogene c-Myc is related to every aspect of the oncogenic process of tumor cells and the alternative activation of macrophages. Hence, we constructed a glycolipid nanocarrier containing ROS-responsive peroxalate linkages (CSOPOSA) for ROS-triggered release of drugs and further modified it with Ex 26 (Ex 26-CSOPOSA), a selective sphingosine 1-phosphate receptor 1 (S1PR1) antagonist, to achieve the dual-targeted delivery of the c-Myc inhibitor JQ1 via S1PR1, which is overexpressed on both tumor cells and TAMs, thereby inducing apoptosis of tumor cells, and blocking M2 polarization of macrophages. More strikingly, our studies found that JQ1 could effectively inhibit the migration of tumor cells induced by M2 macrophages-derived exosomes via blocking Caveolin-1 related intercellular exosome exchange through lncRNA H19 and miR-107. The in vivo results revealed that this dual-targeted delivery strategy effectively inhibited tumor growth and metastasis with less systemic toxicity, providing a potential method for effective tumor treatment.


Assuntos
Caveolina 1 , Exossomos , Neoplasias Experimentais/tratamento farmacológico , Espécies Reativas de Oxigênio , Animais , Linhagem Celular Tumoral , Portadores de Fármacos , Liberação Controlada de Fármacos , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-myc , Células RAW 264.7 , Microambiente Tumoral
8.
Biomaterials ; 275: 120927, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34119887

RESUMO

Early antitumor therapy is an important determinant of survival in patients with cancer. Utilization of specific pathological states, such as hypoxia, greatly promotes the development of intelligent drug delivery systems (DDSs) for targeted antitumor therapy. However, a slight decrease in oxygen levels in early-stage tumors is not sufficient to trigger hypoxia-responsive drug release. Nitroreductase (NTR) is overexpressed in bioreductive hypoxic cancers, and its expression level has been verified to be directly related to hypoxic status. Herein, using glucose oxidase (GOx) as an O2-consuming agent to exacerbate hypoxia, a cascade strategy of GOx-induced overexpression of NTR and amplified NTR-catalyzed release was proposed for early antitumor therapy. Briefly, NTR-sensitive p-nitrobenzyl chloroformate (PNZ-Cl) was adopted to conjugate with the polysaccharide chitosan (CS) and self-assemble into CS-PNZ-Cl micelles. These polymer micelles possess the dual abilities to specifically immobilize GOx and load mitoxantrone (MIT) to form the NTR-responsive nanocascade reactor GOx/MIT@CS-PNZ-Cl. First, as a "key", tumor hypoxia triggers the initial release of GOx, which serves as the O2-consuming agent when catalyzing its reaction with glucose, which is accompanied by H2O2 production. Depleted oxygen levels facilitate the expression of NTR, which in turn amplifies the capacity of the nanocascade reactor to decompose into secondary micelles for enhanced intratumoral permeation. GOx-inspired NTR amplification further elicits MIT release, realizing a synergistic "domino effect" cascade. In addition, upregulated H2O2 has been shown to effectively reverse GSH-mediated MIT resistance, reaching the superior tumor inhibition rate of 93.08%. This GOx-based NTR-responsive nanocascade reactor provides amplification of the bioreductive hypoxic tumor microenvironment for early antitumor therapy.


Assuntos
Glucose Oxidase , Peróxido de Hidrogênio , Neoplasias/tratamento farmacológico , Liberação Controlada de Fármacos , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Glucose Oxidase/metabolismo , Humanos , Nitrorredutases/genética
9.
Mol Ther Nucleic Acids ; 24: 127-139, 2021 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-33738144

RESUMO

Hepatitis B (HB) is a viral infectious disease that seriously endangers human health, and since there are no radical drugs to counter this, effective and safe therapies urgently need to be developed. HB virus (HBV) mainly infects hepatocytes (HCs), while the drugs are easily phagocytosed by Kupffer cells (KCs). In this study, the glutathione concentration difference between HCs and KCs was examined and utilized in an ideal drug-release strategy. Here, galactosylated chitosan-oligosaccharide-SS-octadecylamine (Gal-CSSO) was prepared to accurately deliver 10-23 DNAzyme DrzBC (blocking HBeAg expression) or DrzBS (blocking HBsAg expression) in targeted HB therapy. In vitro Gal-CSSO systems exhibited low cytotoxicity, endosomal escape, and glutathione responsiveness. The HBeAg and HBsAg secretion of HepG2.2.15 was significantly decreased by Gal-CSSO systems, and the maximum inhibition rates were 1.82-fold and 2.38-fold greater than those of commercial Lipofectamine 2000 (Lipo2000) systems. In vivo Gal-CSSO systems exhibited HC targeting and HC microenvironmental responsiveness without noticeable hepatotoxicity or systemic toxicity. The HBeAg and HBsAg titers of the HBV-infected mice were evidently decreased by Gal-CSSO systems, and the inhibition rates were 1.52-fold and 1.22-fold greater than those of Lipo2000 systems. This study presents a kind of glycolipid-like polymer micelles that promise efficient and safe gene therapy of HB.

10.
Biomater Sci ; 8(21): 5955-5968, 2020 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-32966382

RESUMO

The active targeting strategy has achieved inspiring progress for drug accumulation in tumor therapy; however, the insufficient expression level of many potential receptors poses challenges for drug delivery. Poly-γ-glutamic acid (γ-pGluA), a naturally occurring anionic biopolymer, showed high affinity with tumor-associated gamma-glutamyl transpeptidase (GGT), which localized on the cell surface and exhibited intracellular redox homeostasis-dependent expression pattern; thus, GGT was utilized for mediating endocytosis of nanoparticles. Herein, GGT-targeting nanopolyplexes (γ-pGluA-CSO@Fe3+, PCFN) consisting of cationic chitosan and GGT-targeting γ-pGluA blended with iron ion were constructed to load reactive oxygen species-induced menadione (MA) and doxorubicin, which were utilized to investigate the mechanism of GGT up-regulation. Briefly, the pretreated PCFN/MA induced an intracellular oxidative stress environment, which facilitated adjusted up-regulated GGT expression and boosted tumor targeting. Subsequently, the destroyed redox homeostasis sensitized tumors for synergistic therapy. The innovative strategy of augmenting active targeting by disturbing intracellular redox homeostasis offers insight for the application of γ-pGluA-derived nanopolyplexes.


Assuntos
Ácido Glutâmico , gama-Glutamiltransferase , Homeostase , Oxirredução , Regulação para Cima , gama-Glutamiltransferase/genética , gama-Glutamiltransferase/metabolismo
11.
J Zhejiang Univ Sci B ; 21(3): 218-233, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32133799

RESUMO

Metastasis is one of the main reasons causing death in cancer patients. It was reported that chemotherapy might induce metastasis. In order to uncover the mechanism of chemotherapy-induced metastasis and find solutions to inhibit treatment-induced metastasis, the relationship between epithelial-mesenchymal transition (EMT) and doxorubicin (DOX) treatment was investigated and a redox-sensitive small interfering RNA (siRNA) delivery system was designed. DOX-related reactive oxygen species (ROS) were found to be responsible for the invasiveness of tumor cells in vitro, causing enhanced EMT and cytoskeleton reconstruction regulated by Ras-related C3 botulinum toxin substrate 1 (RAC1). In order to decrease RAC1, a redox-sensitive glycolipid drug delivery system (chitosan-ss-stearylamine conjugate (CSO-ss-SA)) was designed to carry siRNA, forming a gene delivery system (CSO-ss-SA/siRNA) downregulating RAC1. CSO-ss-SA/siRNA exhibited an enhanced redox sensitivity compared to nonresponsive complexes in 10 mmol/L glutathione (GSH) and showed a significant safety. CSO-ss-SA/siRNA could effectively transmit siRNA into tumor cells, reducing the expression of RAC1 protein by 38.2% and decreasing the number of tumor-induced invasion cells by 42.5%. When combined with DOX, CSO-ss-SA/siRNA remarkably inhibited the chemotherapy-induced EMT in vivo and enhanced therapeutic efficiency. The present study indicates that RAC1 protein is a key regulator of chemotherapy-induced EMT and CSO-ss-SA/siRNA silencing RAC1 could efficiently decrease the tumor metastasis risk after chemotherapy.


Assuntos
Antineoplásicos/efeitos adversos , Neoplasias da Mama/patologia , Doxorrubicina/efeitos adversos , Sistemas de Liberação de Medicamentos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , RNA Interferente Pequeno/administração & dosagem , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Aminas/química , Quitosana/química , Doxorrubicina/administração & dosagem , Feminino , Humanos , Células MCF-7 , Metástase Neoplásica/prevenção & controle , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Proteínas rac1 de Ligação ao GTP/fisiologia
12.
Biomaterials ; 237: 119793, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32044521

RESUMO

Glioblastoma (GBM) is one of the malignant tumors with high mortality, and the presence of the blood brain barrier (BBB) severely limits the penetration and tissue accumulation of therapeutic agents in the lesion of GBM. Active targeting nanotechnologies can achieve efficient drug delivery in the brain, while still have a very low success rate. Here we revealed a previously unexplored phenomenon that chemotherapy with active targeting nanotechnologies causes pathological BBB functional recovery through VEGF-PI3K-AKT signaling pathway inhibition, accompanied with up-regulated expression of Claudin-5 and Occludin. Seriously, pathological BBB functional recovery induces a significant decrease of intracerebral active targeting nanotechnologies transport during GBM multiple administration, leading to chemotherapy failure in GBM therapeutics. To address this issue, we chose AKT agonist SC79 to transiently re-open functional recovering pathological BBB for continuously intracerebral delivery of brain targeted nanotherapeutics, finally producing an observable anti-GBM effect in vivo, which may offer new sight for other CNS disease treatment.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Nanopartículas , Barreira Hematoencefálica , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Humanos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...