Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nanomedicine ; 20: 102009, 2019 08.
Article in English | MEDLINE | ID: mdl-31085344

ABSTRACT

The emergence and widespread distribution of multi-drug resistant bacteria are considered as a major public health concern. The inabilities to curb severe infections due to antibiotic resistance have increased healthcare costs as well as patient morbidity and mortality. Bacterial biofilms formed by drug-resistant bacteria add additional challenges to treatment. This study describes a solgel based nanoparticle system loaded with garlic extract (GE-np) that exhibits: i) slow and sustained release of garlic components; ii) stabilization of the active components; and iii) significant enhancement of antimicrobial and antibiofilm activity relative to the free garlic extract. Also, GE-np were efficient in penetrating and disrupting the well-established methicillin-resistant Staphylococcus aureus (MRSA) biofilms. Overall, the study suggests that GE-np might be a promising candidate for the treatment of chronic infections due to biofilm forming drug-resistant bacteria.


Subject(s)
Biofilms/drug effects , Garlic/chemistry , Nanoparticles/chemistry , Plant Extracts/pharmacology , Anti-Bacterial Agents/pharmacology , Disulfides , Microbial Sensitivity Tests , Sulfinic Acids/pharmacology
2.
J Drugs Dermatol ; 14(7): 726-32, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26151790

ABSTRACT

BACKGROUND: The treatment of cutaneous wounds in the clinical setting continues to be a clinical challenge and economic burden, with burn wounds being especially formidable. Direct mechanical injury coupled with the transfer of thermal energy leads to tissue necrosis, pro-inflammatory cytokine release and the eventual expansion of an initial wound. Our current therapeutic armamentarium falls short of options to help prevent wound expansion, and therefore new modalities are required. Nitrosating substances such as RSNOs have been proven to be effective in promoting wound closure due to their ability to modulate inflammation, cytokine production and vascular function. OBJECTIVE: We aim to evaluate the efficacy of n-actetylcysteine s-nitrosothiol nanoparticles (NAC-SNO-np) on thermal burn wounds and associated expansion. METHODS: A multi-burn model was utilized to induce three burn wounds on the dorsal surface of BALB/c mice, allowing for evaluation of the burn itself and peripheral tissue. Wounds were excised and processed for histology and immunohistochemistry on day 7 following wounding. RESULTS: Following treatment with NAC-SNO-np, burn wound expansion was attenuated and wound healing was accelerated. Histological analysis revealed increased collagen deposition as well as increased macrophage and decreased neutrophil infiltration into the wound bed. CONCLUSION: NAC-SNO-np represents a platform that harnesses the nitrosative properties of NAC-SNO in order to accelerate the transition from inflammatory to proliferative wound healing. Further studies are needed in order to translate to the clinical setting.


Subject(s)
Acetylcysteine/therapeutic use , Burns/drug therapy , Nanoparticles/therapeutic use , S-Nitrosothiols/therapeutic use , Wound Healing/drug effects , Acetylcysteine/administration & dosage , Administration, Cutaneous , Animals , Disease Models, Animal , Mice , Mice, Inbred BALB C , S-Nitrosothiols/administration & dosage
3.
Nanomedicine ; 11(2): 283-91, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25461287

ABSTRACT

Nitric oxide (NO), an essential agent of the innate immune system, exhibits multi-mechanistic antimicrobial activity. Previously, NO-releasing nanoparticles (NO-np) demonstrated increased antimicrobial activity when combined with glutathione (GSH) due to formation of S-nitrosoglutathione (GSNO), a transnitrosylating agent. To capitalize on this finding, we incorporated the thiol-containing ACE-inhibitor, captopril, with NO-np to form SNO-CAP-np, nanoparticles that both release NO and form S-nitrosocaptopril. In the presence of GSH, SNO-CAP-np demonstrated increased transnitrosylation activity compared to NO-np, as exhibited by increased GSNO formation. Escherichia coli and methicillin-resistant Staphylococcus aureus were highly susceptible to SNO-CAP-np in a dose-dependent fashion, with E. coli being most susceptible, and SNO-CAP-np were nontoxic in zebrafish embryos at translatable concentrations. Given SNO-CAP-np's increased transnitrosylation activity and increased E. coli susceptibility compared to NO-np, transnitrosylation rather than free NO is likely responsible for overcoming E. coli's resistance mechanisms and ultimately killing the pathogen. FROM THE CLINICAL EDITOR: This team of authors incorporated the thiol-containing ACE-inhibitor, captopril, into a nitric oxide releasing nanoparticle system, generating nanoparticles that both release NO and form S-nitrosocaptopril, with pronounced toxic effects on MRSA and E. coli in the presented model system.


Subject(s)
Immune System/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Nanoparticles/administration & dosage , Nitric Oxide/administration & dosage , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/chemistry , Captopril/administration & dosage , Captopril/analogs & derivatives , Captopril/chemistry , Escherichia coli/drug effects , Escherichia coli/pathogenicity , Glutathione/metabolism , Humans , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Nanoparticles/chemistry , Nitric Oxide/chemistry , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
4.
J Sex Med ; 11(12): 2903-14, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25302850

ABSTRACT

INTRODUCTION: Patients undergoing radical prostatectomy (RP) suffer from erectile dysfunction (ED) refractory to phosphodiesterase 5 inhibitors, which act downstream of cavernous nerve (CN)-mediated release of nitric oxide (NO). Direct delivery of NO to the penis could potentially circumvent this limitation. AIM: This study aimed to determine if topically applied NO-releasing nanoparticles (NO-NPs) could elicit erections in a rat model of RP through increased blood flow. METHODS: Twenty-six Sprague Dawley rats underwent bilateral transection of the CN. One week later, NO-NPs were applied topically to the penile shaft in dimethylsulfoxide (DMSO) gel (10 animals) or coconut oil (6 animals). Control animals were treated with empty NPs. Erectile function was determined through the intracorporal pressure/blood pressure ratio (ICP/BP). The effect of the NO-NPs on blood flow was determined using a hamster dorsal window chamber. MAIN OUTCOME MEASURES: Animals were investigated for spontaneous erections, onset and duration of erectile response, and basal ICP/BP ratio. Microcirculatory blood flow was determined through measurements of arteriolar and venular diameter and red blood cell velocity. RESULTS: Eight of 10 animals treated with NO-NPs suspended in DMSO gel had significant increases in basal ICP/BP, and 6 out of these 10 animals demonstrated spontaneous erections of approximately 1 minute in duration. Time to onset of spontaneous erections ranged from 5 to 37 minutes, and they occurred for at least 45 minutes. Similar results were observed with NO-NPs applied in coconut oil. No erectile response was observed in control animal models treated with empty NPs. The hamster dorsal window chamber experiment demonstrated that NO-NPs applied as a suspension in coconut oil caused a significant increase in the microcirculatory blood flow, sustained over 90 minutes. CONCLUSIONS: Topically applied NO-NPs induced spontaneous erections and increased basal ICP in an animal model of RP. These effects are most likely due to increased microcirculatory blood flow. These characteristics suggest that NO-NPs would be useful in penile rehabilitation of patients following RP.


Subject(s)
Disease Models, Animal , Erectile Dysfunction/drug therapy , Penile Erection/drug effects , Phosphodiesterase 5 Inhibitors/administration & dosage , Administration, Cutaneous , Animals , Blood Flow Velocity/drug effects , Erectile Dysfunction/etiology , Male , Muscle, Smooth/drug effects , Penis/blood supply , Phosphodiesterase 5 Inhibitors/pharmacology , Prostatectomy/adverse effects , Rats , Rats, Sprague-Dawley
5.
Am J Emerg Med ; 31(1): 54-63, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22867830

ABSTRACT

BACKGROUND: Plasma expander (PE) based on polyethylene glycol (PEG) conjugated to albumin has shown positive results maintaining blood volume during hemodilution and restoring blood volume during resuscitation from hemorrhagic shock. Polyethylene glycol conjugation to human serum albumin (HSA), PEG-HSA, increases size, weight, and colloidal osmotic pressure, with minor effects on solution viscosity. METHODS: This study was designed to test the hypothesis that PEG-HSA (2 g/dL) produced by direct PEGylation chemistry improves cardiac function during 2 experimental models, (i) moderate hemodilution and (ii) resuscitation from hemorrhagic shock, compared with a conventional colloidal PE (Dextran 70 kd [Dx70], 6 g/dL). Cardiac function was studied using a miniaturized pressure volume conductance catheter implanted in the left ventricle and evaluated in terms of cardiac indices derived from the pressure volume measurements. RESULTS: Polyethylene glycol-HSA increased cardiac output, stroke volume, and stroke work and decreased systemic vascular resistance compared with Dx70 in both experimental models. The improvements induced by PEG-HSA in cardiac function were sustained over the observation time. Polyethylene glycol-HSA cardiac mechanoenergetics changes are the result of increased energy transferred per stroke and decreased resistance of the vasculature connecting the heart. In summary, PEG-HSA decreased left ventricle ejection impedance. CONCLUSION: Ejection of blood diluted with PEG-HSA presented a reduced load to the heart, increased contractile function, and lowered the energy consumed per unit volume compared with Dx70. Our results emphasize the importance of heart function as a parameter to be included in the evaluation changes induced by new PEs.


Subject(s)
Blood Volume/drug effects , Cardiac Output/drug effects , Hemodilution/methods , Plasma Substitutes/pharmacology , Polyethylene Glycols/pharmacology , Serum Albumin/pharmacology , Shock, Hemorrhagic/drug therapy , Ventricular Function, Left/drug effects , Analysis of Variance , Animals , Cardiac Catheterization , Cricetinae , Male , Mesocricetus , Resuscitation/methods , Vascular Resistance/drug effects
6.
Fungal Biol ; 127(7-8): 1224-1230, 2023.
Article in English | MEDLINE | ID: mdl-37495312

ABSTRACT

Fungal diseases are a leading threat to human health, especially in individuals with compromised immunity. Although there have been recent important advances in antifungal drug development, antifungal resistance, drug-drug interactions and difficulties in delivery remain major challenges. Among its pleiotropic actions, nitric oxide (NO) is a key molecule in host defense. We have developed a flexible nanoparticle platform that delivers sustained release of NO and have demonstrated the platform's efficacy against diverse bacteria as well as some fungal species. In this work, we investigate the effects of two NO-releasing particles against a panel of important human yeast. Our results demonstrate that the compounds are both effective against diverse yeast, including ascomycota and basidiomycota species, and that NO-releasing particles may be a potent addition to our armamentarium for the treatment of focal and disseminated mycoses.


Subject(s)
Antifungal Agents , Mycoses , Humans , Antifungal Agents/pharmacology , Nitric Oxide , Saccharomyces cerevisiae , Mycoses/microbiology
7.
Nitric Oxide ; 27(3): 150-60, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22705913

ABSTRACT

Interest in the development of nitric oxide (NO) based therapeutics has grown exponentially due to its well elucidated and established biological functions. In line with this surge, S-nitroso thiol (RSNO) therapeutics are also receiving more attention in recent years both as potential stable sources of NO as well as for their ability to serve as S-nitrosating agents; S-nitrosation of protein thiols is implicated in many physiological processes. We describe two hydrogel based RSNO containing nanoparticle platforms. In one platform the SNO groups are covalently attached to the particles (SNO-np) and the other contains S-nitroso-N-acetyl cysteine encapsulated within the particles (NAC-SNO-np). Both platforms function as vehicles for sustained activity as trans-S-nitrosating agents. NAC-SNO-np exhibited higher efficiency for generating GSNO from GSH and maintained higher levels of GSNO concentration for longer time (24 h) as compared to SNO-np as well as a previously characterized nitric oxide releasing platform, NO-np (nitric oxide releasing nanoparticles). In vivo, intravenous infusion of the NAC-SNO-np and NO-np resulted in sustained decreases in mean arterial pressure, though NAC-SNO-np induced longer vasodilatory effects as compared to the NO-np. Serum chemistries following infusion demonstrated no toxicity in both treatment groups. Together, these data suggest that the NAC-SNO-np represents a novel means to both study the biologic effects of nitrosothiols and effectively capitalize on its therapeutic potential.


Subject(s)
Acetylcysteine/analogs & derivatives , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Vasodilation/drug effects , Acetylcysteine/administration & dosage , Acetylcysteine/chemistry , Animals , Blood Pressure/drug effects , Carbon Dioxide/blood , Cricetinae , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Glutathione/metabolism , Heart Rate/drug effects , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Male , Mesocricetus , Oxygen/blood , S-Nitrosoglutathione/metabolism , S-Nitrosothiols/chemistry , S-Nitrosothiols/pharmacology
8.
J Drugs Dermatol ; 11(12): 1471-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23377518

ABSTRACT

Pseudomonas aeruginosa is a community-acquired, nosocomial pathogen that is an important cause of human morbidity and mortality; it is intrinsically resistant to several antibiotics and is capable of developing resistance to newly developed drugs via a variety of mechanisms. P aeruginosa's ubiquity and multidrug resistance (MDR) warrants the development of innovative methods that overcome its ability to develop resistance. We have previously described a nitric oxide-releasing nanoparticle (NO-np) platform that effectively kills gram-positive and gram-negative organisms in vitro and accelerates clinical recovery in vivo in murine wound and abscess infection models. We have also demonstrated that when glutathione (GSH) is added to NO-np, the nitroso intermediate S-nitrosoglutathione (GSNO) is formed, which has greater activity against P aeruginosa and other gram-negative organisms compared with NO-np alone. In the current study, we evaluate the potential of NO-np to generate GSNO both in vitro and in vivo in a murine excisional wound model infected with an MDR clinical isolate of P aeruginosa. Whereas NO-np alone inhibited P aeruginosa growth in vitro for up to 8 hours, NO-np+GSH completely inhibited P aeruginosa growth for 24 hours. Percent survival in the NO-np+GSH-treated isolates was significantly lower than in the NO-np (36.1% vs 8.3%; P=.004). In addition, NO-np+GSH accelerated wound closure in P aeruginosa-infected wounds, and NO-np+GSH-treated wounds had significantly lower bacterial burden when compared to NO-np-treated wounds (P<.001). We conclude that GSNO is easily generated from our NO-np platform and has the potential to be used as an antimicrobial agent against MDR organisms such as P aeruginosa.


Subject(s)
Nitric Oxide/therapeutic use , Pseudomonas Infections/drug therapy , S-Nitrosoglutathione/metabolism , Surgical Wound Infection/drug therapy , Vasodilator Agents/therapeutic use , Animals , Colony Count, Microbial , Drug Resistance, Multiple, Bacterial , Mice , Mice, Inbred BALB C , Nanoparticles , Nitric Oxide/administration & dosage , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Skin/microbiology , Surgical Wound Infection/microbiology , Vasodilator Agents/administration & dosage , Wound Healing/drug effects
9.
Nanomedicine ; 8(8): 1364-71, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22406184

ABSTRACT

Wound healing is a complex process, coordinated by various biological factors. In immunocompromised states wound healing can be interrupted as a result of decreased numbers of immune cells, impairing the production of effector molecules such as nitric oxide (NO). Therefore, topical NO-releasing platforms, such as diethylenetriamine (DETA NONOate), have been investigated to enhance wound healing. Recently, we demonstrated a nanoparticle platform that releases NO (NO-NPs) in a sustained manner, accelerating wound healing in both uninfected and infected murine wound models. Here, NO-NPs were investigated and compared to DETA NONOate in an immunocompromised wound model using non-obese, diabetic, severe combined immunodeficiency mice. NO-NP treatment accelerated wound closure as compared to controls and DETA NONOate treatment. In addition, histological assessment revealed that wounds treated with NO-NPs had less inflammation, more collagen deposition, and more blood vessel formation as compared to other groups, consistent with our previous data in immunocompetent animals. These data suggest that NO-NPs may serve as a novel wound-healing therapy in the setting of immunocompromised states associated with impaired wound healing. FROM THE CLINICAL EDITOR: Wound healing in an immunocompromised host is often incomplete and is a source of major concern in such conditions. This work demonstrates in a murine model that in these settings NO releasing nanoparticles significantly enhance wound healing.


Subject(s)
Nanoparticles , Nitric Oxide , Wound Healing/drug effects , Animals , Collagen/metabolism , Female , Mice , Mice, Inbred NOD , Mice, SCID , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nitric Oxide/administration & dosage , Nitric Oxide/chemistry , Platelet Aggregation/drug effects , Skin/drug effects , Skin/pathology , Wound Infection/drug therapy , Wound Infection/pathology
10.
Am J Physiol Heart Circ Physiol ; 300(1): H49-56, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21057038

ABSTRACT

Erythrocyte free hemoglobin (Hb) induces vasoconstriction due to nitric oxide (NO) scavenging, limiting the NO available for vascular smooth muscle. The central objective of this study was to restore NO bioavailability using long-lived circulating NO-releasing nanoparticles (NO-np) to reverse the vasoconstriction and hypertension induced by polymerized bovine Hb (PBH) NO scavenging. PBH (13 g/dl) was infused in a volume equal to 10% of the animal blood volume. Intravascular NO supplementation was provided with an infusion of NO-np (10 and 20 mg/kg body wt). This study was performed using the hamster window chamber model to concurrently access systemic and microvascular hemodynamics. Infusion of PBH increased blood pressure and induced vasoconstriction. Treatment with 10 and 20 mg/kg NO-np reduced the blood pressure and vasoconstriction induced by PBH. Moreover, the higher dose of NO-np decreased blood pressure and induced vasodilation compared with baseline, respectively. Treatment with NO-np to decrease PBH-induced vasoconstriction increased methemoglobin levels and plasma nitrite and nitrate. In conclusion, NO-np counteracted both systemic hypertension and decreased the vasoconstrictor effects of PBH infusion, improving systemic and microvascular function. Based on the observed physiological properties, NO-np has clear potential as a therapeutic agent to replenish NO in situations where NO production is impaired, insufficient, or consumed, thereby preventing vascular complications.


Subject(s)
Hemoglobins/administration & dosage , Nitric Oxide/administration & dosage , Vasoconstriction/drug effects , Analysis of Variance , Animals , Blood Pressure/drug effects , Cricetinae , Heart Rate/drug effects , Hemoglobins/metabolism , Male , Mesocricetus , Microcirculation/drug effects , Nanoparticles , Nitric Oxide/metabolism , Random Allocation , Vasoconstriction/physiology
11.
Nitric Oxide ; 25(4): 381-6, 2011 Nov 30.
Article in English | MEDLINE | ID: mdl-21946032

ABSTRACT

Nitric oxide (NO) plays a vital role in mammalian host defense through a variety of mechanisms. In particular, NO can oxidize to form reactive nitrogen species or interact with protein thiols and metal centers, blocking essential microbial processes. S-nitrosoglutathione (GSNO), a potent NO donor formed by the interaction of NO with intracellular glutathione (GSH), is a major factor in this pathway and is considered one of the strongest naturally occurring nitrosating agent. We previously described the broad-spectrum antimicrobial activity of a nanoparticulate platform capable of controlled and sustained release of NO (NO-np). Interestingly, in vivo efficacy of the NO-np surpassed in vitro data generated. We hypothesized that the enhanced activity was in part achieved via the interaction between the generated NO and available GSH, forming GSNO. In the current study, we investigated the efficiency of NO-np to form GSNO in the presence of GSH was evaluated, and assessed the antimicrobial activity of the formed GSNO against methicillin resistant Staphylococcus aureus (MRSA), Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa. When GSH was combined with NO-np, GSNO was rapidly produced and significant concentrations of GSNO were maintained for >24h. The GSNO generated was more effective compared to NO-np alone against all bacterial strains examined, with P. aeruginosa being the most sensitive and K. pneumoniae the most resistant. We conclude that the combination of NO-np with GSH is an effective means of generating GSNO, and presents a novel approach to potent antimicrobial therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Nanoparticles/chemistry , Nitric Oxide/chemistry , S-Nitrosoglutathione/pharmacology , Escherichia coli/drug effects , Glutathione/chemistry , Klebsiella pneumoniae/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Nitric Oxide/pharmacology , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacology , Pseudomonas aeruginosa/drug effects , S-Nitrosoglutathione/chemistry
12.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33872220

ABSTRACT

The microtubule (MT) cytoskeleton plays a critical role in axon growth and guidance. Here, we identify the MT-severing enzyme fidgetin-like 2 (FL2) as a negative regulator of axon regeneration and a therapeutic target for promoting nerve regeneration after injury. Genetic knockout of FL2 in cultured adult dorsal root ganglion neurons resulted in longer axons and attenuated growth cone retraction in response to inhibitory molecules. Given the axonal growth-promoting effects of FL2 depletion in vitro, we tested whether FL2 could be targeted to promote regeneration in a rodent model of cavernous nerve (CN) injury. The CNs are parasympathetic nerves that regulate blood flow to the penis, which are commonly damaged during radical prostatectomy (RP), resulting in erectile dysfunction (ED). Application of FL2-siRNA after CN injury significantly enhanced functional nerve recovery. Remarkably, following bilateral nerve transection, visible and functional nerve regeneration was observed in 7 out of 8 animals treated with FL2-siRNA, while no control-treated animals exhibited regeneration. These studies identify FL2 as a promising therapeutic target for enhancing regeneration after peripheral nerve injury and for mitigating neurogenic ED after RP - a condition for which, at present, only poor treatment options exist.


Subject(s)
ATPases Associated with Diverse Cellular Activities/physiology , Axon Guidance/genetics , Axons/metabolism , Ganglia, Spinal/cytology , Microtubule-Associated Proteins/physiology , Nerve Regeneration/genetics , Neurons/metabolism , Peripheral Nerve Injuries/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Animals , Cells, Cultured , Male , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubules , Penis/innervation , Prostatectomy , RNA Interference , RNA, Small Interfering
13.
Transl Vis Sci Technol ; 10(1): 17, 2021 01.
Article in English | MEDLINE | ID: mdl-33510956

ABSTRACT

Purpose: The purpose of this study was to determine the efficacy of nanoparticle-encapsulated Fidgetin-like 2 (FL2) siRNA (FL2-NPsi), a novel therapeutic agent targeting the FL2 gene, for the treatment of corneal alkaline chemical injury. Methods: Eighty 12-week-old, male Sprague-Dawley rats were divided evenly into 8 treatment groups: prednisolone, empty nanoparticles, control-NPsi (1 µM, 10 µM, and 20 µM) and FL2-NPsi (1 µM, 10 µM, and 20 µM). An alkaline burn was induced onto the cornea of each rat, which was then treated for 14 days according to group assignment. Clinical, histopathologic, and immunohistochemical analyses were conducted to assess for wound healing. FL2-NPsi-mediated knockdown of FL2 was confirmed by in vitro quantitative polymerase chain reaction (qPCR). Toxicity assays were performed to assess for apoptosis (terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling [TUNEL] assay) and nerve damage (whole mount immunochemical staining). Statistical analyses were performed using Student's t-test and ANOVA. Results: Compared with controls, FL2-NPsi-treated groups demonstrated enhanced corneal wound healing, with the 10 and 20 µM FL2-NPsi-treated groups demonstrating maximum rates of corneal re-epithelialization as assessed by ImageJ software, enhanced corneal transparency, and improved stromal organization on histology. Immunohistochemical analysis of vascular endothelial cells, macrophages, and neutrophils did not show significant differences between treatment groups. FL2-NPsi was not found to be toxic to nerves or induce apoptosis (p = 0.917). Conclusions: Dose-response studies found both 10 and 20 µM FL2-NPsi to be efficacious in this rat model. FL2-NPsi may offer a novel treatment for corneal alkaline chemical injuries. Translational Relevance: Basic cell biology findings about the microtubule cytoskeleton were used to design a therapeutic to enhance corneal cell migration, highlighting the promise of targeting microtubules to regulate corneal wound healing.


Subject(s)
Endothelial Cells , Eye Burns , Animals , Cornea , Eye Burns/chemically induced , Male , Microtubules , Rats , Rats, Sprague-Dawley
14.
J Fungi (Basel) ; 6(2)2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32545506

ABSTRACT

Candida auris (C. auris) is an emerging pathogenic fungal species that is especially worrisome due to its high mortality rates and widespread antifungal resistance. Previous studies have demonstrated the efficacy of nitric oxide (NO) nanoparticles on Candida species, and, to our knowledge, this is the first study to investigate the antifungal effects of a NO-generating nanoparticle on C. auris. Six C. auris strains were incubated with a nanoparticle (NAC-SNO-np), which releases N-acetylcysteine S-nitrosothiol (NAC-SNO) and N-acetylcysteine (NAC), and generates NO, through colony forming unit (CFU) assays, and confocal laser scanning microscopy. NAC-SNO-np effectively eradicates planktonic and biofilm C. auris. Across all six strains, 10 mg/mL NAC-SNO-np significantly reduced the number of CFUs (p < 0.05) and demonstrated a >70% decrease in biofilm viability (p < 0.05). NAC-SNO-np effectively eradicates planktonic C. auris and significantly reduces C. auris biofilm formation. Hence, this novel NO-releasing nanoparticle shows promise as a future therapeutic.

15.
Int Forum Allergy Rhinol ; 10(2): 223-232, 2020 02.
Article in English | MEDLINE | ID: mdl-31834677

ABSTRACT

BACKGROUND: Bacterial biofilms are implicated in the pathogenesis of chronic rhinosinusitis. Nitric oxide (NO) is a key immune effector with potent antimicrobial effects, but a short half-life limits achievement of therapeutic concentrations. We hypothesized that manuka honey (MH) could induce sustained reduction of nitrite to NO causing biofilm disruption and that this effect would be enhanced with the addition of a NO-releasing microparticle. METHODS: Porous organosilica microparticles containing nitrosylated thiol groups were formulated (SNO-MP). MH was combined with serial dilutions of nitrite. NO release was evaluated using a NO analyzer. The susceptibility of 2 strains of Pseudomonas aeruginosa biofilms to these NO-releasing platforms was evaluated using confocal microscopy. Cell viability and biofilm volume were quantified. Statistical analysis was performed using the Mann-Whitney U test with SPSS software. RESULTS: MH with nitrite generated a linear increase in NO formation. SNO-MP induced a bolus release of NO within 5 minutes, followed by a sustained plateau phase. MH with nitrite combined with SNO-MP enhanced NO release during the plateau phase. MH with nitrite reduced biofilm live cells and volume by 88.5% to 96.9% and 95.1% to 95.6%, respectively, vs control (p < 0.0001). SNO-MP reduced live cells and volume by 61.0% to 98.5% and 74.7% to 85.7%, respectively, vs control (p < 0.0001). MH with nitrite combined with SNO-MP nearly eradicated biofilm, with a 98.3% to 99.8% (log 1.8-2.6) reduction in viability and a 91.4% to 97.7% decrease in volume (p < 0.0001 vs control). CONCLUSION: A novel platform that generates NO using MH and nitrite produces a potent anti-biofilm effect, which can be further enhanced with the addition of SNO-MP.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Honey , Nitric Oxide/chemistry , Nitrites/administration & dosage , Organosilicon Compounds/administration & dosage , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Chronic Disease , Nitrites/chemistry , Organosilicon Compounds/chemistry , Oxidation-Reduction , Pseudomonas aeruginosa/physiology , Rhinitis/therapy , Sinusitis/therapy
16.
Article in English | MEDLINE | ID: mdl-19922165

ABSTRACT

Plasma expander-like properties of albumin induced on hexa as well as dodecacPEGylation using Extension Arm Facilitated PEGylation platform make it an excellent resuscitation fluid. PEGylation induced changes in the structure, drug binding, and plasma expander-like properties of bovine serum albumin has been now investigated as a function of PEGylation. The molecular volume of albumin increases on PEGylation nearly linearly; in the beginning up to about six PEG chains are conjugated, then plateau off, while the viscosity and colloidal osmotic pressure change very little initially and then increase exponentially as a function of PEG chains conjugated. PEGylation has essentially no influence on the secondary structure or drug properties of albumin. Tryphtophyl fluorescence of albumin is quenched on PEGylation as a direct correlate of the changes in molecular radius of PEG-albumin. It is concluded that hexaPEGylated and dodecaPEGylated albumin belong to two different configurational states of PEG-albumin in terms of packing of PEG-chains on the molecular surface of the protein. The results suggest a transition of PEGylated albumin from the initial mushroom-like conformation to brush conformation as the PEGylation increases. The therapeutic efficacy of the two PEGylated species is needed to establish the optimum level of PEGylation to function as resuscitation fluids.


Subject(s)
Polyethylene Glycols/chemistry , Serum Albumin/chemistry , Serum Albumin/therapeutic use , Animals , Cattle , Molecular Conformation , Osmotic Pressure , Protein Conformation , Resuscitation/methods , Viscosity
17.
Anal Biochem ; 374(2): 231-42, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18158909

ABSTRACT

The design of the extension arm-facilitated PEGylation (EAFP) of proteins takes advantage of the high selective and quantitative aspects of the thiol-maleimide reaction. However, the efficiency of EAFP with hemoglobin varied with the batches of maleimide-PEG. The low level of functionalization of monomethoxy-PEG (mPEG) as maleimide-PEG has been now investigated as the potential source of this variation. New chemical approaches for the estimation of the functionalization of mPEG using the reaction of the thiol groups of glutathione, dithiothreitol, and hemoglobin with maleimide-PEG have been developed. The single-step modular approach to the synthesis of maleimidophenyl-PEG (MPPEG) that involved the condensation of p-maleimidophenyl isocyanate with mPEG has been optimized to generate a product with an overall purity of 80%. The NMR approach correlates well with the estimates made by the new chemical approaches. Commercial maleimide-PEG reagents synthesized using multiple steps exhibited a lower level of functionalization as reflected by these chemical estimations. The better functionalization of MPPEG increases the efficiency of EAFP as reflected by the generation of hexaPEGylated Hb and the masking of the D antigen of RBCs. This new EAFP protocol is expected to improve the cost effectiveness of the generation of hexaPEGylated Hb, PEGylated albumin, and PEGylated RBCs as new PEGylated therapeutics.


Subject(s)
Maleimides/chemistry , Polyethylene Glycols/chemistry , Blood Group Antigens/analysis , Dithiothreitol/chemical synthesis , Dithiothreitol/chemistry , Erythrocytes/chemistry , Glutathione/chemistry , Hemoglobin A/analysis , Hemoglobin A/chemistry , Indicators and Reagents/chemistry , Magnetic Resonance Spectroscopy
18.
Artif Cells Nanomed Biotechnol ; 45(2): 297-304, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26924283

ABSTRACT

Curcumin is an effective and safe anticancer agent, and also known to induce vasodilation, but its hydrophobicity limits its clinical application. In this study, a simple emulsion method was developed to prepare biodegradable poly (ethylene glycol)-poly (lactic acid) (PEG-PLA) nanospheres to encapsulate curcumin to improve its solubility and stability. The nanoparticle size was around 150 nm with a narrow size distribution. Fluorescence microscopy showed that curcumin encapsulated PEG-PLA nanospheres were taken up rapidly by Hela and MDA-MB-231 cancer cells. This novel nanoparticulate carrier may improve the bioavailability of curcumin without affecting its anticancer properties.


Subject(s)
Curcumin , Drug Delivery Systems/methods , Nanospheres/chemistry , Neoplasms/drug therapy , Polyesters , Polyethylene Glycols , Curcumin/chemistry , Curcumin/pharmacology , HeLa Cells , Humans , Neoplasms/metabolism , Polyesters/chemistry , Polyesters/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology
19.
Sci Rep ; 7(1): 9675, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28852085

ABSTRACT

Microtubule-stabilizing drugs have gained popularity for treating injured adult axons, the rationale being that increased stabilization of microtubules will prevent the axon from retracting and fortify it to grow through inhibitory molecules associated with nerve injury. We have posited that a better approach would be not to stabilize the microtubules, but to increase labile microtubule mass to levels more conducive to axonal growth. Recent work on fetal neurons suggests this can be accomplished using RNA interference to reduce the levels of fidgetin, a microtubule-severing protein. Methods to introduce RNA interference into adult neurons, in vitro or in vivo, have been problematic and not translatable to human patients. Here we show that a novel nanoparticle approach, previously shown to deliver siRNA into tissues and organs, enables siRNA to gain entry into adult rat dorsal root ganglion neurons in culture. Knockdown of fidgetin is partial with this approach, but sufficient to increase the labile microtubule mass of the axon, thereby increasing axonal growth. The increase in axonal growth occurs on both a favorable substrate and a growth-inhibitory molecule associated with scar formation in injured spinal cord. The nanoparticles are readily translatable to in vivo studies on animals and ultimately to clinical applications.


Subject(s)
ATPases Associated with Diverse Cellular Activities/antagonists & inhibitors , Drug Carriers/administration & dosage , Drug Carriers/metabolism , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubules/metabolism , Nanoparticles/metabolism , Neurons/physiology , Neuroprotective Agents/metabolism , RNA, Small Interfering/metabolism , Animals , Cells, Cultured , Nerve Regeneration , Rats
20.
PLoS One ; 12(1): e0170041, 2017.
Article in English | MEDLINE | ID: mdl-28099525

ABSTRACT

Nearly 21 million components of blood and whole blood and transfused annually in the United States, while on average only 13.6 million units of blood are donated. As the demand for Red Blood Cells (RBCs) continues to increase due to the aging population, this deficit will be more significant. Despite decades of research to develop hemoglobin (Hb) based oxygen (O2) carriers (HBOCs) as RBC substitutes, there are no products approved for clinical use. Lumbricus terrestris erythrocruorin (LtEc) is the large acellular O2 carrying protein complex found in the earthworm Lumbricus terrestris. LtEc is an extremely stable protein complex, resistant to autoxidation, and capable of transporting O2 to tissue when transfused into mammals. These characteristics render LtEc a promising candidate for the development of the next generation HBOCs. LtEc has a short half-life in circulation, limiting its application as a bridge over days, until blood became available. Conjugation with polyethylene glycol (PEG-LtEc) can extend LtEc circulation time. This study explores PEG-LtEc pharmacokinetics and pharmacodynamics. To study PEG-LtEc pharmacokinetics, hamsters instrumented with the dorsal window chamber were subjected to a 40% exchange transfusion with 10 g/dL PEG-LtEc or LtEc and followed for 48 hours. To study the vascular response of PEG-LtEc, hamsters instrumented with the dorsal window chamber received multiple infusions of 10 g/dL PEG-LtEc or LtEc solution to increase plasma LtEc concentration to 0.5, then 1.0, and 1.5 g/dL, while monitoring the animals' systemic and microcirculatory parameters. Results confirm that PEGylation of LtEc increases its circulation time, extending the half-life to 70 hours, 4 times longer than that of unPEGylated LtEc. However, PEGylation increased the rate of LtEc oxidation in vivo. Vascular analysis verified that PEG-LtEc showed the absence of microvascular vasoconstriction or systemic hypertension. The molecular size of PEG-LtEc did not change the colloid osmotic pressure or blood volume expansion capacity compared to LtEc, due to LtEc's already large molecular size. Taken together, these results further encourage the development of PEG-LtEc as an O2 carrying therapeutic.


Subject(s)
Hemoglobins/chemistry , Hemoglobins/pharmacokinetics , Oligochaeta/metabolism , Polyethylene Glycols/chemistry , Animals , Arterioles/drug effects , Blood Flow Velocity/drug effects , Blood Substitutes , Capillaries/drug effects , Half-Life , Infusions, Intravenous , Male , Mesocricetus , Nitrite Reductases/chemistry , Oxygen/metabolism , Venules/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL