Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Chem Commun (Camb) ; 60(74): 10136-10139, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39189125

ABSTRACT

For the first time, the dominant magnetoelectric activity of ZIF-67-derived carbonaceous microparticles embedded with Co nanoparticles and distinctive magnetothermal effect of MIL-88B-derived Fe3O4 nanocubes decorated on carbonaceous microrods, respectively, were explored to be controlled by the structure of the MOF-derived electrically conductive carbonaceous matrix and metal nanoparticles.

2.
J Control Release ; 354: 417-428, 2023 02.
Article in English | MEDLINE | ID: mdl-36627025

ABSTRACT

Fibrosis is an excessive accumulation of extracellular matrix (ECM) that may cause severe organ dysfunction. Nitric oxide (NO), a multifunctional gaseous signaling molecule, may inhibit fibrosis, and delivery of NO may serve as a potential antifibrotic strategy. However, major limitations in the application of NO to treat fibrotic diseases include its nonspecificity, short half-life and low availability in fibrotic tissue. Herein, we aimed to develop a stimuli-responsive drug carrier to deliver NO to halt kidney fibrosis. We manufactured a nanoparticle (NP) composed of pH-sensitive poly[2-(diisopropylamino)ethyl methacrylate (PDPA) polymers to encapsulate a NO donor, a dinitrosyl iron complex (DNIC; [Fe2(µ-SEt)2(NO)4]). The NPs were stable at physiological pH 7.4 but disintegrated at pH 4.0-6.0. The NPs showed significant cytotoxicity to cultured human myofibroblasts and were able to inhibit the activation of myofibroblasts, as indicated by a lower expression level of α-smooth muscle actin and the synthesis of a major ECM component, collagen I, in cultured human myofibroblasts. When given to mice treated with unilateral ureteral ligation/obstruction (UUO) to induce kidney fibrosis, these NPs remained in blood at a stable concentration for as long as 24 h and might enter the fibrotic kidneys to suppress myofibroblast activation and collagen I production, leading to a 70% reduction in the fibrotic area. In summary, our strategy to assemble a NO donor, the iron nitrosyl complex DNIC, into pH-responsive NPs proves effective in treating renal fibrosis and warrants further investigation for its therapeutic potential.


Subject(s)
Kidney Diseases , Ureteral Obstruction , Mice , Humans , Animals , Kidney , Nitric Oxide/metabolism , Kidney Diseases/drug therapy , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Collagen Type I/metabolism , Fibrosis , Hydrogen-Ion Concentration , Mice, Inbred C57BL
3.
J Control Release ; 352: 920-930, 2022 12.
Article in English | MEDLINE | ID: mdl-36334859

ABSTRACT

While immunotherapy has emerged as a promising strategy to treat glioblastoma multiforme (GBM), the limited availability of immunotherapeutic agents in tumors due to the presence of the blood-brain barrier (BBB) and immunosuppressive tumor microenvironment dampens efficacy. Nitric oxide (NO) plays a role in modulating both the BBB and tumor vessels and could thus be delivered to disrupt the BBB and improve the delivery of immunotherapeutics into GBM tumors. Herein, we report an immunotherapeutic approach that utilizes CXCR4-targeted lipid­calcium-phosphate nanoparticles with NO donors (LCP-NO NPs). The delivery of NO resulted in enhanced BBB permeability and thus improved gene delivery across the BBB. CXCR4-targeted LCP-NO NPs delivered siRNA against the immune checkpoint ligand PD-L1 to GBM tumors, silenced PD-L1 expression, increased cytotoxic T cell infiltration and activation in GBM tumors, and suppressed GBM progression. Thus, the codelivery of NO and PD-L1 siRNA by these CXCR4-targeted NPs may serve as a potential immunotherapy for GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Nanoparticles , Humans , Glioblastoma/drug therapy , B7-H1 Antigen , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Nitric Oxide/therapeutic use , Brain Neoplasms/drug therapy , Immunotherapy , Tumor Microenvironment , Cell Line, Tumor , Receptors, CXCR4/genetics
4.
ACS Appl Mater Interfaces ; 14(3): 3849-3863, 2022 Jan 26.
Article in English | MEDLINE | ID: mdl-35019259

ABSTRACT

Nitric oxide (NO) is an endogenous gasotransmitter regulating alternative physiological processes in the cardiovascular system. To achieve translational application of NO, continued efforts are made on the development of orally active NO prodrugs for long-term treatment of chronic cardiovascular diseases. Herein, immobilization of NO-delivery [Fe2(µ-SCH2CH2COOH)2(NO)4] (DNIC-2) onto MIL-88B, a metal-organic framework (MOF) consisting of biocompatible Fe3+ and 1,4-benzenedicarboxylate (BDC), was performed to prepare a DNIC@MOF microrod for enhanced oral delivery of NO. In simulated gastric fluid, protonation of the BDC linker in DNIC@MOF initiates its transformation into a DNIC@tMOF microrod, which consisted of DNIC-2 well dispersed and confined within the BDC-based framework. Moreover, subsequent deprotonation of the BDC-based framework in DNIC@tMOF under simulated intestinal conditions promotes the release of DNIC-2 and NO. Of importance, this discovery of transformer-like DNIC@MOF provides a parallel insight into its stepwise transformation into DNIC@tMOF in the stomach followed by subsequent conversion into molecular DNIC-2 in the small intestine and release of NO in the bloodstream of mice. In comparison with acid-sensitive DNIC-2, oral administration of DNIC@MOF results in a 2.2-fold increase in the oral bioavailability of NO to 65.7% in mice and an effective reduction of systolic blood pressure (SBP) to a ΔSBP of 60.9 ± 4.7 mmHg in spontaneously hypertensive rats for 12 h.


Subject(s)
Biocompatible Materials/pharmacology , Metal-Organic Frameworks/pharmacology , Nitric Oxide/chemistry , Prodrugs/pharmacology , Administration, Oral , Animals , Biocompatible Materials/administration & dosage , Blood Pressure/drug effects , Electrodes , Hydrogen-Ion Concentration , Materials Testing , Metal-Organic Frameworks/administration & dosage , Mice , Nitric Oxide/administration & dosage , Particle Size , Prodrugs/chemistry , Surface Properties
5.
ACS Appl Mater Interfaces ; 14(5): 6343-6357, 2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35080366

ABSTRACT

Nitric oxide (NO) is an essential endogenous signaling molecule regulating multifaceted physiological functions in the (cardio)vascular, neuronal, and immune systems. Due to the short half-life and location-/concentration-dependent physiological function of NO, translational application of NO as a novel therapeutic approach, however, awaits a strategy for spatiotemporal control on the delivery of NO. Inspired by the magnetic hyperthermia and magneto-triggered drug release featured by Fe3O4 conjugates, in this study, we aim to develop a magnetic responsive NO-release material (MagNORM) featuring dual NO-release phases, namely, burst and steady release, for the selective activation of NO-related physiology and treatment of bacteria-infected cutaneous wound. After conjugation of NO-delivery [Fe(µ-S-thioglycerol)(NO)2]2 with a metal-organic framework (MOF)-derived porous Fe3O4@C, encapsulation of obtained conjugates within the thermo-responsive poly(lactic-co-glycolic acid) (PLGA) microsphere completes the assembly of MagNORM. Through continuous/pulsatile/no application of the alternating magnetic field (AMF) to MagNORM, moreover, burst/intermittent/slow release of NO from MagNORM demonstrates the AMF as an ON/OFF switch for temporal control on the delivery of NO. Under continuous application of the AMF, in particular, burst release of NO from MagNORM triggers an effective anti-bacterial activity against both Gram-positive Staphylococcus aureus (S. aureus) and Gram-negative Escherichia coli (E. coli). In addition to the magneto-triggered bactericidal effect of MagNORM against E. coli-infected cutaneous wound in mice, of importance, steady release of NO from MagNORM without the AMF promotes the subsequent collagen formation and wound healing in mice.


Subject(s)
Ferrosoferric Oxide/chemistry , Magnetic Fields , Metal-Organic Frameworks/chemistry , Microspheres , Nitric Oxide/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Drug Carriers/chemistry , Escherichia coli/drug effects , Escherichia coli/physiology , Escherichia coli Infections/drug therapy , Male , Mice , Mice, Inbred BALB C , Nitric Oxide/pharmacology , Nitric Oxide/therapeutic use , Skin/microbiology , Skin/pathology , Staphylococcus aureus/drug effects , Wound Healing/drug effects
6.
Dalton Trans ; 48(26): 9431-9453, 2019 Jul 02.
Article in English | MEDLINE | ID: mdl-30990502

ABSTRACT

Iron, the most abundant transition metal ion in humans, participates in the biosynthesis, translocation, signal transduction, and transformation of nitric oxide through its encapsulation in the form of heme, [Fe-S], and [Fe(NO)2] cofactors within a variety of enzymes and proteins. After the review on nitric oxide synthase (NOS) and soluble guanylate cyclase (sGC) for the biosynthesis and detection of NO, in this report, we discuss the natural utilization of the [Fe(NO)2] motif for translocation of endogenous NO and the translational development of synthetic dinitrosyl iron complexes (DNICs) for biomedical applications. A mechanistic study of NO-release and NO-transfer reactivity of structure-characterized DNICs promoted the discovery of cell-penetrating and in vivo NO-delivery reactivity for treatment of cancer and wound healing in diabetes. Beyond activation of sGC and vasodilation, phase I/II clinical trials of glutathione-bound DNICs (Oxacom®) against hypertension encourage bioinorganic engineering of DNICs into scaffolds for tissue regeneration and repair relying on anti-bacterial, anti-inflammation, cytoprotective, and proliferative effects of NO.


Subject(s)
Iron/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Nitrogen Oxides/chemistry , Nitrogen Oxides/metabolism , Tissue Engineering , Humans , Iron/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL