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.
ACS Appl Bio Mater ; 7(3): 1435-1440, 2024 03 18.
Article in English | MEDLINE | ID: mdl-38447089

ABSTRACT

We report the design of a blood-contacting glucose monitor with a nitric oxide (NO)-releasing metal-organic framework (MOF) embedded within the outer polymer layer of a glucose sensor to promote the release of NO from endogenous NO donors. The sensors were tested by using amperometry across a range of glucose concentrations to assess whether the presence of either the MOF or NO decreased the performance of the glucose monitor. Even though signal response was diminished, the sensors maintained a good regression fit (R2 = 0.9944) and a similar dynamic range and reproducibility in the presence of S-nitrosoglutathione.


Subject(s)
Metal-Organic Frameworks , Nitric Oxide , Reproducibility of Results , Nitric Oxide Donors , Glucose
2.
ACS Biomater Sci Eng ; 8(8): 3438-3449, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35776832

ABSTRACT

Medical devices that require substantial contact between blood and a foreign surface would be dramatically safer if constructed from materials that prevent clot formation and coagulation disturbance at the blood-biomaterial interface. Nitric oxide (NO), an endogenous inhibitor of platelet activation in the vascular endothelium, could provide anticoagulation at the blood-surface interface when applied to biomaterials. We investigated an application of a copper-based metal-organic framework, H3[(Cu4Cl)3(BTTri)8-(H2O)12]·72H2O where H3BTTri = 1,3,5-tris(1H-1,2,3-triazole-5-yl)benzene] (CuBTTri), which has been shown to be an effective catalyst to generate NO from S-nitrosothiols that are endogenously present in blood. A method was developed to apply a CuBTTri composite coating to Tygon medical tubing used for extracorporeal lung support devices. The stability and activity of the coating were evaluated during 72 h dynamic saline flow testing (1.5-2.5 L/min, n = 3) with scanning electron microscopy imaging and inductively coupled mass-spectroscopy analysis. Compatibility of the coating with whole blood was assessed with a panel of hemocompatibility tests during 6 h circulation of swine donor blood in an ex vivo circulation loop constructed with CuBTTri tubing or unmodified Tygon (1.5 L/min blood flow rate, n = 8/group). Thrombus deposition and catalytic activity of the CuBTTri tubing were assessed following blood exposure. The coating remained stable during 72 h saline flow experiments at clinically relevant flow rates. No adverse effects were observed relative to controls during blood compatibility testing, to include no significant changes in platelet count (p = 0.42), platelet activation indicated by P-selectin expression (p = 0.57), coagulation panel values, or methemoglobin fraction (p = 0.18) over the 6 h circulation period. CuBTTri within the coating generated NO following blood exposure in the presence of biologically relevant concentrations of an NO donor. CuBTTri composite coating was stable and blood compatible in this pilot study and requires further investigation of efficacy using in vivo models conducted with clinically relevant blood flow rates and study duration.


Subject(s)
Metal-Organic Frameworks , Thrombosis , Animals , Biocompatible Materials , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitric Oxide Donors , Pilot Projects , Swine , Thrombosis/prevention & control
3.
ACS Appl Mater Interfaces ; 13(48): 56931-56943, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34818503

ABSTRACT

It has been previously demonstrated that metal nanoparticles embedded into polymeric materials doped with nitric oxide (NO) donor compounds can accelerate the release rate of NO for therapeutic applications. Despite the advantages of elevated NO surface flux for eradicating opportunistic bacteria in the initial hours of application, metal nanoparticles can often trigger a secondary biocidal effect through leaching that can lead to unfavorable cytotoxic responses from host cells. Alternatively, copper-based metal organic frameworks (MOFs) have been shown to stabilize Cu2+/1+ via coordination while demonstrating longer-term catalytic performance compared to their salt counterparts. Herein, the practical application of MOFs in NO-releasing polymeric substrates with an embedded NO donor compound was investigated for the first time. By developing composite thermoplastic silicon polycarbonate polyurethane (TSPCU) scaffolds, the catalytic effects achievable via intrapolymeric interactions between an MOF and NO donor compound were investigated using the water-stable copper-based MOF H3[(Cu4Cl)3(BTTri)8-(H2O)12]·72H2O (CuBTTri) and the NO donor S-nitroso-N-acetyl-penicillamine (SNAP). By creating a multifunctional triple-layered composite scaffold with CuBTTri and SNAP, the surface flux of NO from catalyzed SNAP decomposition was found tunable based on the variable weight percent CuBTTri incorporation. The tunable NO surface fluxes were found to elicit different cytotoxic responses in human cell lines, enabling application-specific tailoring. Challenging the TSPCU-NO-MOF composites against 24 h bacterial growth models, the enhanced NO release was found to elicit over 99% reduction in adhered and over 95% reduction in planktonic methicillin-resistant Staphylococcus aureus, with similar results observed for Escherichia coli. These results indicate that the combination of embedded MOFs and NO donors can be used as a highly efficacious tool for the early prevention of biofilm formation on medical devices.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biomimetic Materials/pharmacology , Metal-Organic Frameworks/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Catalysis , Cells, Cultured , Copper/chemistry , Humans , Materials Testing , Microbial Sensitivity Tests , Molecular Conformation , Nitric Oxide Donors/chemical synthesis , Nitric Oxide Donors/chemistry , Particle Size , Surface Properties
4.
Adv Healthc Mater ; 10(7): e2001748, 2021 04.
Article in English | MEDLINE | ID: mdl-33448158

ABSTRACT

When flowing whole blood contacts medical device surfaces, the most common blood-material interactions result in coagulation, inflammation, and infection. Many new blood-contacting biomaterials have been proposed based on strategies that address just one of these common modes of failure. This study proposes to mitigate unfavorable biological reactions that occur with blood-contacting medical devices by designing multifunctional surfaces, with features optimized to meet multiple performance criteria. These multifunctional surfaces incorporate the release of the small molecule hormone nitric oxide (NO) with surface chemistry and nanotopography that mimic features of the vascular endothelial glycocalyx. These multifunctional surfaces have features that interact with coagulation components, inflammatory cells, and bacterial cells. While a single surface feature alone may not be sufficient to achieve multiple functions, the release of NO from the surfaces along with their modification to mimic the endothelial glycocalyx synergistically improves platelet-, leukocyte-, and bacteria-surface interactions. This work demonstrates that new blood-compatible materials should be designed with multiple features, to better address the multiple modes of failure of blood-contacting medical devices.


Subject(s)
Biocompatible Materials , Endothelium, Vascular , Biomimetics , Blood Coagulation , Blood Platelets , Surface Properties
5.
Nitric Oxide ; 92: 18-25, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31398487

ABSTRACT

S-Nitrosothiols (RSNOs) such as S-nitrosoglutathione (GSNO) and S-nitroso-N-acetylpenicillamine (SNAP) are susceptible to decomposition by stimuli including heat, light, and trace metal ions. Using stepwise isothermal thermogravimetric analysis (TGA), we observed that NO-forming homolytic cleavage of the S-N bond occurs at 134.7 ±â€¯0.8 °C in GSNO and 132.8 ±â€¯0.9 °C in SNAP, contrasting with the value of 150 °C that has been previously reported for both RSNOs. Using mass spectrometry (MS), nuclear magnetic resonance (NMR), and attenuated total reflectance Fourier transform infrared spectroscopy (ATR-FTIR), we analyzed the decomposition products from TGA experiments. The organic product of GSNO decomposition was glutathione disulfide, while SNAP decomposed to form N-acetylpenicillamine disulfide as well as other products, including tri- and tetrasulfides. In addition, we assessed the relative solution stabilities of GSNO and SNAP under common laboratory conditions, which include variable temperature, pH, and light exposure with rigorous exclusion of trace metal ions by chelation. GSNO exhibited greater stability than SNAP over a 7-day period except in one instance. Both RSNOs demonstrated an inverse relationship between solution stability and temperature, with refrigeration considerably extending shelf life. A decrease in pH from 7.4 to 5.0 also enhanced the stability of both RSNOs. A further decrease in pH from 5.0 to 3.0 resulted in decreased stability for both RSNOs, and is notably the only occasion in which SNAP proved more stable than GSNO. After 1 h of exposure to overhead fluorescent lighting, both RSNOs displayed high susceptibility to light-induced decomposition. After 7 h, GSNO and SNAP decomposed 19.3 ±â€¯0.5% and 30 ±â€¯2%, respectively.


Subject(s)
S-Nitroso-N-Acetylpenicillamine/chemistry , S-Nitrosoglutathione/chemistry , Molecular Structure , Proton Magnetic Resonance Spectroscopy , Thermogravimetry
6.
Nitric Oxide ; 84: 16-21, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30630054

ABSTRACT

S-Nitrosothiols (RSNOs) such as S-nitrosoglutathione (GSNO) are known to produce nitric oxide (NO) through thermal, photolytic, and metal ion-promoted pathways, which has led to their increasing use as exogenous sources of therapeutic NO. Despite the burgeoning NO release applications for RSNOs, their susceptibility to metal-promoted decomposition has rarely been examined in a uniform manner through the specific measurement of NO release. In this study, the ability of various transition and post-transition metal ions to promote NO release from GSNO was surveyed by chemiluminescence-based NO detection. Substantial NO formation (>10-fold increase relative to GSNO baseline) was detected after the addition of Cu2+, Au3+, Pd2+, Pt2+, and V3+. Modest increases were observed in the cases of Co2+, Hf4+, Fe2+, Fe3+, Mn2+, Hg2+, Ni2+, Ag+, Sn2+, and Zr4+, while no effect was evident for Al3+, Cr3+, Pb2+, Sc3+, and Zn2+. It was further observed that In+ compounds initiate the apparent NO-forming decomposition of GSNO, while In0 and In3+ are inactive, indicating that In+ exerts a previously unknown effect on GSNO.


Subject(s)
Indium/chemistry , Nitric Oxide Donors/chemistry , Nitric Oxide/chemical synthesis , S-Nitrosoglutathione/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL