Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Nitric Oxide ; 150: 37-46, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39038732

ABSTRACT

The combination of nitric oxide (NO) donors with nanomaterials has emerged as a promising approach to reduce postharvest losses. The encapsulation of NO donors provides protection from rapid degradation and controlled release, enhancing the NO effectiveness in postharvest treatments. Moreover, the application method can also influence postharvest responses. In this study, two application methods were evaluated, spraying and immersion, using S-nitrosoglutathione (GSNO, a NO donor) in free and encapsulated forms on papaya fruit. Our hypothesis was that GSNO encapsulated in chitosan nanoparticles would outperform the free form in delaying fruit senescence. In addition, this study marks the pioneering characterization of chitosan nanoparticles containing GSNO within the framework of a postharvest investigation. Overall, our findings indicate that applying encapsulated GSNO (GSNO-NP-S) through spraying preserves the quality of papaya fruit during storage. This method not only minimizes weight loss, ethylene production, and softening, but also stimulates antioxidant responses, thereby mitigating oxidative damage. Consequently, it stands out as the promising technique for delaying papaya fruit senescence. This innovative approach holds the potential to enhance postharvest practices and advance sustainable agriculture.


Subject(s)
Carica , Chitosan , Fruit , Nitric Oxide Donors , S-Nitrosoglutathione , Carica/chemistry , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemistry , Fruit/chemistry , S-Nitrosoglutathione/pharmacology , S-Nitrosoglutathione/chemistry , Chitosan/chemistry , Chitosan/pharmacology , Oxidative Stress/drug effects , Nanoparticles/chemistry , Food Preservation/methods
2.
J Tissue Eng Regen Med ; 14(6): 807-818, 2020 06.
Article in English | MEDLINE | ID: mdl-32330363

ABSTRACT

Mounting evidence showing that local nitric oxide (NO) delivery may significantly improve the wound healing process has stimulated the development of wound dressings capable of releasing NO topically. Herein, we describe the preparation of a self-expandable NO-releasing hydrolyzed collagen sponge (CS), charged with the endogenously found NO donor, S-nitrosoglutathione (GSNO). We show that cold pressed and GSNO-charged CS (CS/GSNO) undergo self-expansion to its original 3D shape upon water absorption to a swelling degree of 2,300 wt%, triggering the release of free NO. Topical application of compressed CS/GSNO on wounds in an animal model showed that exudate absorption by CS/GSNO leads to the release of higher NO doses during the inflammatory phase and progressively lower NO doses at later stages of the healing process. Moreover, treated animals showed significant increase in the mRNA expression levels of monocyte chemoattractant protein-1 (MCP-1), murine macrophage marker (F4/80), transforming growth factor beta (TGF-ß), stromal cell-derived factor 1 (SDF-1), insulin-like growth factor-1 (IGF-1), nitric oxide synthase(iNOS), and matrix metalloproteinase(MMP-9). Cluster differentiation 31 (CD31), vascular endothelial growth factor (VEGF), and F4/80 were measured on Days 7 and 12 by immunohistochemistry in the cicatricial tissue. These results indicate that the topical delivery of NO enhances the migration and infiltration of leucocytes, macrophages, and keratinocytes to the wounded tissue, as well as the neovascularization and collagen deposition, which are correlated with an accelerated wound closure. Thus, self-expandable CS/GSNO may represent a novel biocompatible and active wound dress for the topical delivery of NO on wounds.


Subject(s)
Collagen , Nitric Oxide , S-Nitrosoglutathione , Wound Healing/drug effects , Wounds and Injuries , Animals , Collagen/chemistry , Collagen/pharmacology , Disease Models, Animal , Drug Implants/chemistry , Drug Implants/pharmacokinetics , Drug Implants/pharmacology , Male , Mice , Nitric Oxide/chemistry , Nitric Oxide/pharmacokinetics , Nitric Oxide/pharmacology , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/pharmacokinetics , S-Nitrosoglutathione/pharmacology , Wounds and Injuries/drug therapy , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
3.
Nitric Oxide ; 94: 108-113, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31759127

ABSTRACT

Nitric oxide (NO) is a crucial molecule in the human body. The encapsulation of exogenous NO donors into chitosan nanoparticles (CS NPs) has been widely used to overcome NO drawbacks in pharmacological applications, such as, its short half-life. The NO donor, S-nitrosoglutathione (GSNO), was encapsulated into CS NPs (GSNO-CS NPs) and characterized by AFM and DLS measurements. The nanoparticles presented a hydrodynamic size of 123.3 ± 1.5 nm and a polydispersity of 0.25 ± 0.01. The ability of GSNO-CS NPs, combined with UV irradiation, to deliver NO was evaluated using ex vivo human skin. The human skin was pre-treated with GSNO-CS NPs, in the presence and absence of UV irradiation. The results showed that the combined treatment significantly increased the NO and S-nitrosothiol levels in human skin. This effect can emulate the cardiovascular benefits related to NO without negative side effects of skin exposure to UV light.


Subject(s)
Chitosan/chemistry , Nanoparticles/chemistry , Nitric Oxide Donors/chemistry , Nitric Oxide/pharmacology , S-Nitrosoglutathione/chemistry , Skin/drug effects , Humans , Hydrodynamics , Nitric Oxide/chemistry , Particle Size , Surface Properties , Ultraviolet Rays
4.
J Colloid Interface Sci ; 544: 217-229, 2019 May 15.
Article in English | MEDLINE | ID: mdl-30849619

ABSTRACT

Poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) (PEO-PPO-PEO) (F127) hydrogels have been used to deliver nitric oxide (NO) topically in biomedical applications. Here, the effect of F127 microenvironments on the photochemical NO release from S-nitrosoglutathione (GSNO) was investigated in F127 solutions 7.6 wt% 15 wt% and 22.5 wt% at 15 °C and 37 °C. Small-angle X-ray Scattering (SAXS) and Differential Scanning Calorimetry (DSC) measurements, along with proton Nuclear Magnetic Resonance (1H NMR) spectral shifts and T2 relaxation data at six different concentration-temperature conditions, allowed identifying F127 microphases characterized by: a sol phase of unimers; micelles in non-defined periodic order, and a gel phase of cubic packed micelles. Kinetic measurements showed that GSNO photodecompositon proceeds faster in micellized F127 where GSNO is segregated to the intermicellar microenvironment. Real time kinetic monitoring of NO release and T2 relaxation profiles showed that NO is preferentially partitioned into the hydrophobic PPO cores of the F127 micelles, with the consequent decrease in its rate of release to the gas phase. These results show that F127 microphases affect both the kinetics of GSNO photodecomposition and the rate of NO escape and can be used to modulate the photochemical NO delivery from F127/GSNO solutions.


Subject(s)
Hydrogels/chemistry , Nitric Oxide/chemistry , Poloxamer/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Propylene Glycols/chemistry , S-Nitrosoglutathione/chemistry , Drug Carriers/chemistry , Drug Liberation , Kinetics , Micelles , Photochemical Processes , Temperature
5.
Nitric Oxide ; 84: 30-37, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30630056

ABSTRACT

Primary S-nitrosothiols (RSNOs) have received significant attention for their ability to modulate NO signaling in many physiological and pathophysiological processes. Such actions and their potential pharmaceutical uses demand a better knowledge of their stability in aqueous solutions. Herein, we investigated the effects of concentration, temperature, pH, room light and metal ions on the long-term kinetic behavior of two representative primary RSNOs, S-nitrosoglutathione (GSNO) and S-nitroso-N-acetylcysteine (SNAC). The thermal decomposition of GSNO and SNAC were shown to be affected by the auto-catalytic action of the thiyl radicals. At 25 °C in the dark and protected from the catalytic action of metal ions, GSNO and SNAC solutions 1 mM showed half-lives of 49 and 76 days, and apparent activation energies of 84 ±â€¯14 and 90 ±â€¯6 kJ mol-1, respectively. Both GSNO and SNAC exhibited increased stability in the pH range 5-7. At high pH the decomposition pathway of GSNO involves the formation of an intermediate (GS-NO22-), which decomposes generating GSH and nitrite. GSNO solutions displayed lower sensitivity to the catalytic action of metal ions than SNAC and the exposure to room light led to a 5-fold increase in the initial rates of decomposition of both RSNOs. In all comparisons, SNAC solutions showed higher stability than GSNO solutions. These findings provide strategic information about the stability of GSNO and SNAC and may open new perspectives for their use as experimental or therapeutic NO donors.


Subject(s)
Acetylcysteine/analogs & derivatives , S-Nitrosoglutathione/chemistry , Acetylcysteine/chemical synthesis , Acetylcysteine/chemistry , Hydrogen-Ion Concentration , Kinetics , Light , S-Nitrosoglutathione/chemical synthesis , Temperature
6.
Acta Biomater ; 74: 312-325, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29777958

ABSTRACT

Topical nitric oxide (NO) delivery has been shown to accelerate wound healing. However, delivering NO to wounds at appropriate rates and doses requires new biomaterial-based strategies. Here, we describe the development of supramolecular interpolymer complex hydrogels comprising PEO-PPO-PEO (F127) micelles embedded in a poly(acrylic acid) (PAA) matrix, with S-nitrosoglutathione (GSNO) molecules dissolved in the hydrophilic domain. We show that PAA:F127/GSNO hydrogels start releasing NO upon hydration at rates controlled by their rates of water absorption. SAXS measurements indicate that the supramolecular structure of the hydrogels retains long-range order domains of F127 micelles. The PAA/F1227 hydrogels displayed dense morphologies and reduced rates of hydration. The NO release rates remain constant over the first 200 min, are directly correlated with the hydration rates of the PAA:F127/GSNO hydrogels, and can be modulated in the range of 40 nmol/g h to 1.5 µmol/g h by changing the PAA:F127 mass ratio. Long-term NO-release profiles over 5 days are governed by the first-order exponential decay of GSNO, with half-lives in the range of 0.5-3.4 days. A preliminary in vivo study on full-thickness excisional wounds in mice showed that topical NO release from the PAA:F127/GSNO hydrogels is triggered by exudate absorption and leads to increased angiogenesis and collagen fiber organization, as well as TGF-ß, IGF-1, SDF-1, and IL-10 gene expressions in the cicatricial tissue. In summary, these results suggest that hydration-controlled NO release from topical PAA:F127/GSNO hydrogels is a potential strategy for enhancing wound healing. STATEMENT OF SIGNIFICANCE: The topical delivery of nitric oxide (NO) to wounds may provide significant beneficial results and represent a promising strategy to treat chronic wounds. However, wound dressings capable of releasing NO after application and allowing the modulation of NO release rates, demand new platforms. Here, we describe a novel strategy to overcome these challenges, based on the use of supramolecular poly(acrylic acid) (PAA):F127 hydrogels charged with the NO donor S-nitrosoglutathione (GSNO) from whereby the NO release can be triggered by exudate absorption and delivered to the wound at rates controlled by the PAA:F127 mass ratio. Preliminary in vivo results offer a proof of concept for this strategy by demonstrating increased angiogenesis; collagen fibers organization; and TGF-ß, IGF-1, SDF-1, and IL-10 gene expressions in the cicatricial tissue after topical treatment with a PAA:F127/GSNO hydrogel.


Subject(s)
Acrylic Resins , Hydrogels , Nitric Oxide , Polyethylenes , Polypropylenes , Wound Healing/drug effects , Wounds and Injuries , Acrylic Resins/pharmacokinetics , Acrylic Resins/pharmacology , Animals , Cytokines/biosynthesis , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Gene Expression Regulation/drug effects , Hydrogels/chemistry , Hydrogels/pharmacokinetics , Hydrogels/pharmacology , Mice , Micelles , Nitric Oxide/chemistry , Nitric Oxide/pharmacokinetics , Nitric Oxide/pharmacology , Polyethylenes/chemistry , Polyethylenes/pharmacokinetics , Polyethylenes/pharmacology , Polypropylenes/chemistry , Polypropylenes/pharmacokinetics , Polypropylenes/pharmacology , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/pharmacokinetics , S-Nitrosoglutathione/pharmacology , Wounds and Injuries/drug therapy , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
7.
Colloids Surf B Biointerfaces ; 130: 182-91, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25907598

ABSTRACT

Nitric oxide (NO) releasing biomaterials represent a potential strategy for use as active wound dressings capable of accelerating wound healing. Topical NO-releasing poly(vinyl alcohol) (PVA) films and Pluronic F127 hydrogels (F127) have already exhibited effective skin vasodilation and wound healing actions. In this study, we functionalized PVA films with SNO groups via esterification with a mixture of mercaptosucinic acid (MSA) and thiolactic acid (TLA) followed by S-nitrosation of the SH moieties. These films were combined with an underlying layer of poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide), i.e., PEO-PPO-PEO (Pluronic F127) hydrogel and used for the topical treatment of skin lesions in an animal model. The mixed esterification of PVA with MSA and TLA led to chemically crosslinked PVA-SNO films with a high swelling capacity capable of spontaneously releasing NO. Real time NO-release measurements revealed that the hydrogel layer reduces the initial NO burst from the PVA-SNO films. We demonstrate that the combination of PVA-SNO films with F127 hydrogel accelerates wound contraction, decreases wound gap and cellular density and accelerates the inflammatory phase of the lesion. These results were reflected in an increase in myofibroblastic differentiation and collagen type III expression in the cicatricial tissue. Therefore, PVA-SNO films combined with F127 hydrogel may represent a new approach for active wound dressings capable of accelerating wound healing.


Subject(s)
Hydrogels/chemistry , Nitric Oxide/chemistry , Poloxamer/chemistry , Polyvinyl Alcohol/chemistry , Actins/metabolism , Animals , Antigens, Differentiation/metabolism , Blotting, Western , Hydrogels/metabolism , Hydrogels/pharmacology , Immunohistochemistry , Male , Mice , Nitric Oxide/metabolism , Poloxamer/metabolism , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Polyvinyl Alcohol/metabolism , Polyvinyl Alcohol/pharmacology , Propylene Glycols/chemistry , Propylene Glycols/metabolism , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/metabolism , Skin/metabolism , Skin/pathology , Skin/physiopathology , Sulfhydryl Compounds/chemistry , Thiomalates/chemistry , Time Factors , Wound Healing/drug effects
8.
Colloids Surf B Biointerfaces ; 108: 178-84, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23545088

ABSTRACT

Clinical complications of implantable polypropylene (PP) meshes used to repair urinary incontinence and vaginal prolapse may be associated with their low surface energy and consequent poor tissue integration. For improving tissue integration, we impregnated monofilament PP meshes with physically crosslinked poly(vinyl alcohol) (PVA), resulting in PVA deposits tightly attached inside the knot spaces of the PP knit. While preserving the mesh porosity, the PVA deposits acted as an array of hydrophilic regions leading to a great increase in the overall mesh wettability, reflected by a contact angle decrease from 111 to ca. 66°. The PVA deposits were also used as reservoirs for the local release of S-nitrosoglutathione (GSNO), a nitric oxide (NO) donor. Plain and impregnated PP meshes (1.0cm×1.0cm) were implanted in the subcutaneous tissue of 21 adult female Wistar rats. Histological analysis of the abdominal wall 21 days after the surgeries revealed lower edema and greater angiogenesis while a marked decrement of NOx concentration in the tissue surrounding the impregnated meshes was observed after 2 days. These results indicate that PVA and PVA/GSNO impregnation might be a new strategy for decreasing the frequency of mesh extrusion after PP mesh implants.


Subject(s)
Abdominal Wall/blood supply , Biocompatible Materials/chemistry , Nitric Oxide Donors/chemistry , Polypropylenes/chemistry , Polyvinyl Alcohol/chemistry , S-Nitrosoglutathione/chemistry , Abdominal Wall/surgery , Animals , Biocompatible Materials/pharmacology , Female , Microscopy, Electron, Scanning , Neovascularization, Physiologic , Nitric Oxide/metabolism , Polyvinyl Alcohol/pharmacokinetics , Rats , Rats, Wistar , S-Nitrosoglutathione/pharmacokinetics , Surgical Mesh , Wettability
9.
Nitric Oxide ; 11(3): 263-72, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15566973

ABSTRACT

Incorporation of nitric oxide (NO) donors in non-toxic polymeric matrices can be a useful strategy for allowing topical NO delivery. We have incorporated the NO-donor S-nitrosoglutathione (GSNO) into a liquid poly(ethylene glycol) (PEG)/H2O matrix through the S-nitrosation of GSH by a NO/O2 gas mixture. Kinetic measurements of GSNO decomposition associated with NO release were performed at 25, 35, and 45 degrees C in the dark and under irradiation with UV/Vis light, lambda>480 nm and lambda=333 nm. NO release from the liquid matrix to the gas phase was confirmed by mass spectrometry. The PEG/H2O matrix stabilizes GSNO leading to expressive reductions in the initial rates of thermal and photochemical NO release, compared to aqueous GSNO solution. This matrix effect is assigned to diffusional constrains imposed on the escape of the NO and GS radicals formed in the solvent cage. This effect allows the storage of PEG-GSNO formulations for extended periods (more than 65 days at freezer) with negligible decomposition. PEG-GSNO formulation seems therefore to be applicable in topical NO delivery and GSNO displays potential as a percutaneous absorption enhancer. Moreover, the rate of NO release can be locally increased by irradiation with visible light.


Subject(s)
Drug Delivery Systems/methods , Nitric Oxide Donors/metabolism , Nitric Oxide/administration & dosage , Nitric Oxide/metabolism , Polyethylene Glycols/chemistry , S-Nitrosoglutathione/metabolism , Administration, Topical , Drug Delivery Systems/instrumentation , Kinetics , Nitric Oxide Donors/chemistry , Photochemistry , S-Nitrosoglutathione/chemistry , Solutions , Spectrum Analysis , Temperature
10.
Braz J Med Biol Res ; 36(5): 587-94, 2003 May.
Article in English | MEDLINE | ID: mdl-12715077

ABSTRACT

The photogeneration of nitric oxide (NO) using laser flash photolysis was investigated for S-nitroso-glutathione (GSNO) and S-nitroso-N-acetylcysteine (NacySNO) at pH 6.4 (PBS/HCl) and 7.4 (PBS). Irradiation of S-nitrosothiol with light (lambda = 355 nm followed by absorption spectroscopy) resulted in the homolytic decomposition of NacySNO and GSNO to generate radicals (GS and NacyS ) and NO. The release of NO from donor compounds measured with an ISO-Nometer apparatus was larger at pH 7.4 than pH 6.4. NacySNO was also incorporated into dipalmitoyl-phosphatidylcholine liposomes in the presence and absence of zinc phthalocyanine (ZnPC), a well-known photosensitizer useful for photodynamic therapy. Liposomes are usually used as carriers for hydrophobic compounds such as ZnPC. Inclusion of ZnPC resulted in a decrease in NO liberation in liposomal medium. However, there was a synergistic action of both photosensitizers and S-nitrosothiols resulting in the formation of other reactive species such as peroxynitrite, which is a potent oxidizing agent. These data show that NO release depends on pH and the medium, as well as on the laser energy applied to the system. Changes in the absorption spectrum were monitored as a function of light exposure.


Subject(s)
Acetylcysteine/analogs & derivatives , Acetylcysteine/chemistry , Indoles/chemistry , Nitric Oxide/chemistry , Organometallic Compounds/chemistry , Photolysis , S-Nitrosoglutathione/chemistry , Isoindoles , Liposomes , Zinc Compounds
SELECTION OF CITATIONS
SEARCH DETAIL