Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 1 de 1
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Nanobiotechnology ; 22(1): 299, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38812031

RÉSUMÉ

BACKGROUND: Discrepancies in the utilization of reactive oxygen species (ROS) between cancer cells and their normal counterparts constitute a pivotal juncture for the precise treatment of cancer, delineating a noteworthy trajectory in the field of targeted therapies. This phenomenon is particularly conspicuous in the domain of nano-drug precision treatment. Despite substantial strides in employing nanoparticles to disrupt ROS for cancer therapy, current strategies continue to grapple with challenges pertaining to efficacy and specificity. One of the primary hurdles lies in the elevated levels of intracellular glutathione (GSH). Presently, predominant methods to mitigate intracellular GSH involve inhibiting its synthesis or promoting GSH efflux. However, a conspicuous gap remains in the absence of a strategy capable of directly and efficiently clearing GSH. METHODS: We initially elucidated the chemical mechanism underpinning oridonin, a diminutive pharmacological agent demonstrated to perturb reactive oxygen species, through its covalent interaction with glutathione. Subsequently, we employed the incorporation of maleimide-liposomes, renowned for their capacity to disrupt the ROS delivery system, to ameliorate the drug's water solubility and pharmacokinetics, thereby enhancing its ROS-disruptive efficacy. In a pursuit to further refine the targeting for acute myeloid leukemia (AML), we harnessed the maleic imide and thiol reaction mechanism, facilitating the coupling of Toll-like receptor 2 (TLR2) peptides to the liposomes' surface via maleic imide. This strategic approach offers a novel method for the precise removal of GSH, and its enhancement endeavors are directed towards fortifying the precision and efficacy of the drug's impact on AML targets. RESULTS: We demonstrated that this peptide-liposome-small molecule machinery targets AML and consequently induces cell apoptosis both in vitro and in vivo through three disparate mechanisms: (I) Oridonin, as a Michael acceptor molecule, inhibits GSH function through covalent bonding, triggering an initial imbalance of oxidative stress. (II) Maleimide further induces GSH exhaustion, aggravating redox imbalance as a complementary augment with oridonin. (III) Peptide targets TLR2, enhances the directivity and enrichment of oridonin within AML cells. CONCLUSION: The rationally designed nanocomplex provides a ROS drug enhancement and targeted delivery platform, representing a potential solution by disrupting redox balance for AML therapy.


Sujet(s)
Diterpènes de type kaurane , Glutathion , Leucémie aigüe myéloïde , Liposomes , Espèces réactives de l'oxygène , Diterpènes de type kaurane/composition chimique , Diterpènes de type kaurane/pharmacologie , Glutathion/métabolisme , Glutathion/composition chimique , Liposomes/composition chimique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/métabolisme , Humains , Espèces réactives de l'oxygène/métabolisme , Animaux , Souris , Lignée cellulaire tumorale , Récepteur de type Toll-2/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Apoptose/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE