logo logo
Functional poly(ε-caprolactone)/chitosan dressings with nitric oxide-releasing property improve wound healing. Zhou Xin,Wang He,Zhang Jimin,Li Xuemei,Wu Yifan,Wei Yongzhen,Ji Shenglu,Kong Deling,Zhao Qiang Acta biomaterialia Wound healing dressings are increasingly needed clinically due to the large number of skin damage annually. Nitric oxide (NO) plays a key role in promoting wound healing, thus biomaterials with NO-releasing property receive increasing attention as ideal wound dressing. In present study, we prepared a novel functional wound dressing by combining electrospun poly(ε-caprolactone) (PCL) nonwoven mat with chitosan-based NO-releasing biomaterials (CS-NO). As-prepared PCL/CS-NO dressing released NO sustainably under the physiological conditions, which was controlled by the catalysis of β-galactosidase. In vivo wound healing characteristics were further evaluated on full-thickness cutaneous wounds in mice. Results showed that PCL/CS-NO wound dressings remarkably accelerated wound healing process through enhancing re-epithelialization and granulation formation and effectively improved the organization of regenerated tissues including epidermal-dermal junction, which could be ascribed to the pro-angiogenesis, immunomodulation, and enhanced collagen synthesis provided by the sustained release of NO. Therefore, PCL/CS-NO may be a promising candidate for wound dressings, especially for the chronic wound caused by the ischemia. STATEMENT OF SIGNIFICANCE:Serious skin damage caused by trauma, surgery, burn or chronic disease has become one of the most serious clinical problems. Therefore, there is an increasing demand for ideal wound dressing that can improve wound healing. Due to the vital role of nitric oxide (NO), we developed a novel functional wound dressing by combining electrospun polycaprolactone (PCL) mat with NO-releasing biomaterial (CS-NO). The sustained release of NO from PCL/CS-NO demonstrated positive effects on wound healing, including pro-angiogenesis, immunomodulation, and enhanced collagen synthesis. Hence, wound healing process was remarkably accelerated and the organization of regenerated tissues was effectively improved as well. Taken together, PCL/CS-NO dressing may be a promising candidate for wound treatment, especially for the chronic wound caused by the ischemia. 10.1016/j.actbio.2017.03.011
Nitric oxide-releasing microparticles as a potent antimicrobial therapeutic against chronic rhinosinusitis bacterial isolates. Abuzeid Waleed M,Girish Vallerinteavide Mavelli,Fastenberg Judd H,Draganski Andrew R,Lee Andrew Y,Nosanchuk Joshua D,Friedman Joel M International forum of allergy & rhinology BACKGROUND:Bacteria, particularly in the biofilm state, may be implicated in the pathogenesis of chronic rhinosinusitis (CRS) and enhance antibiotic resistance. Nitric oxide (NO) is a gaseous immunomodulator with antimicrobial activity and a short half-life, complicating achievement of therapeutic concentrations. We hypothesized that a novel microparticle-based delivery platform, which allows for adjustable release of NO, could exhibit potent antibacterial effects. METHODS:Porous organosilica microparticles (SNO-MP) containing nitrosylated thiol groups were formulated. Dissociation of the nitrosothiol groups generates NO at body temperature. The susceptibility of bacterial isolates from CRS patients to SNO-MP was evaluated through a colony forming unit (CFU) assay. Serial dilutions of SNO-MP in triplicate were incubated with isolates in suspension for 6 hours followed by plating on tryptic soy agar and overnight incubation followed by CFU quantification. Statistical analysis was performed with SPSS using one-way analysis of variance with Bonferroni correction. RESULTS:SNO-MP displayed antibacterial activity against gram-positive (methicillin-resistant and -sensitive Staphylococcus aureus) and gram-negative (Pseudomonas aeruginosa, Enterobacter aerogenes, and Proteus mirabilis) isolates. SNO-MP induced dose-dependent reductions in CFU across all strains. Compared with controls and blank nanoparticles, SNO-MP (10 mg/mL) induced a 99.99%-100% reduction in CFU across all isolates, equivalent to a 5-9 log kill (p < 0.005). There was no statistically significant difference in CFU concentration between controls and blank microparticles. CONCLUSION:SNO-MP demonstrates potent bactericidal effect against antibiotic-resistant CRS bacterial strains. 10.1002/alr.22185
Topical N-acetylcysteine accelerates wound healing in vitro and in vivo via the PKC/Stat3 pathway. Tsai Min-Ling,Huang Hui-Pei,Hsu Jeng-Dong,Lai Yung-Rung,Hsiao Yu-Ping,Lu Fung-Jou,Chang Horng-Rong International journal of molecular sciences N-Acetylcysteine (Nac) is an antioxidant administered in both oral and injectable forms. In this study, we used Nac topically to treat burn wounds in vitro and in vivo to investigate mechanisms of action. In vitro, we monitored glutathione levels, cell proliferation, migration, scratch-wound healing activities and the epithelialization-related proteins, matrixmetalloproteinase-1 (MMP-1) and proteins involved in regulating the expression of MMP-1 in CCD-966SK cells treated with Nac. Various Nac concentrations (0.1, 0.5, and 1.0 mM) increased glutathione levels, cell viability, scratch-wound healing activities and migration abilities of CCD-966SK cells in a dose-dependent manner. The MMP-1 expression of CCD-966SK cells treated with 1.0 mM Nac for 24 h was significantly increased. Levels of phosphatidylinositol 3-kinase (PI3K), protein kinase C (PKC), janus kinase 1 (Jak1), signal transducer and activator of transcription 3 (Stat3), c-Fos and Jun, but not extracellular signal-regulated protein kinases 1 and 2 (Erk1/2), were also significantly increased in a dose-dependent manner compared to the controls. In addition, Nac induced collagenous expression of MMP-1 via the PKC/Stat3 signaling pathway. In vivo, a burn wound healing rat model was applied to assess the stimulation activity and histopathological effects of Nac, with 3.0% Nac-treated wounds being found to show better characteristics on re-epithelialization. Our results demonstrated that Nac can potentially promote wound healing activity, and may be a promising drug to accelerate burn wound healing. 10.3390/ijms15057563
Antibacterial properties of nitric oxide-releasing porous silicon nanoparticles. Hasanzadeh Kafshgari M,Delalat B,Harding F J,Cavallaro A,Mäkilä E,Salonen J,Vasilev K,Voelcker N H Journal of materials chemistry. B In this study, the antibacterial efficacy of NO-releasing porous silicon nanoparticles (pSiNPs) is reported. NO-releasing pSiNPs were produced via the conjugation of S-nitrosothiol (SNO) and S-nitrosoglutathione (GSNO) donors to the nanoparticle surfaces. The release of the conjugated NO caused by the decomposition of the conjugated SNO and GSNO was boosted in the presence of ascorbic acid. The released NO was bactericidal to Gram-positive (Staphylococcus aureus) and Gram-negative bacteria (Escherichia coli), and eliminated bacterial growth within 2 h of incubation without compromising the viability of mammalian cells. These results demonstrate the advantages of NO-releasing pSiNPs for antibacterial applications, for example, in chronic wound treatment. 10.1039/c5tb02551f
N-acetylcysteine S-nitrosothiol Nanoparticles Prevent Wound Expansion and Accelerate Wound Closure in a Murine Burn Model. Landriscina Angelo,Musaev Tagai,Rosen Jamie,Ray Anjana,Nacharaju Parimala,Nosanchuk Joshua D,Friedman Adam J Journal of drugs in dermatology : JDD 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.
Photochemistry of nitric oxide and S-nitrosothiols in human skin. Pelegrino Milena T,Paganotti André,Seabra Amedea B,Weller Richard B Histochemistry and cell biology Nitric oxide (NO) is related to a wide range of physiological processes such as vasodilation, macrophages cytotoxicity and wound healing. The human skin contains NO precursors (NO). Those are mainly composed of nitrite (NO), nitrate (NO), and S-nitrosothiols (RSNOs) which forms a large NO store. These NO stores in human skin can mobilize NO to blood stream upon ultraviolet (UV) light exposure. The main purpose of this study was to evaluate the most effective UV light wavelength to generate NO and compare it to each NO precursor in aqueous solution. In addition, the UV light might change the RSNO content on human skin. First, we irradiated pure aqueous solutions of NO and NO and mixtures of NO and glutathione and NO and S-nitrosoglutathione (GSNO) to identify the NO release profile from those species alone. In sequence, we evaluated the NO generation profile on human skin slices. Human skin was acquired from redundant plastic surgical samples and the NO and RSNO measurements were performed using a selective NO electrochemical sensor. The data showed that UV light could trigger the NO generation in skin with a peak at 280-285 nm (UVB range). We also observed a significant RSNO formation in irradiated human skin, with a peak at 320 nm (UV region) and at 700 nm (visible region). Pre-treatment of the human skin slice using NO and thiol (RSHs) scavengers confirmed the important role of these molecules in RSNO formation. These findings have important implications for clinical trials with potential for new therapies. 10.1007/s00418-020-01858-w
Sustained Nitric Oxide Release from a Tertiary S-Nitrosothiol-based Polyphosphazene Coating. Lutzke Alec,Tapia Jesus B,Neufeld Megan J,Reynolds Melissa M ACS applied materials & interfaces Nitric oxide (NO) occurs naturally in mammalian biochemistry as a critical signaling molecule and exhibits antithrombotic, antibacterial, and wound-healing properties. NO-forming biodegradable polymers have been utilized in the development of antithrombotic or antibacterial materials for biointerfacial applications, including tissue engineering and the fabrication of erodible coatings for medical devices such as stents. Use of such NO-forming polymers has frequently been constrained by short-term release or limited NO storage capacity and has led to the pursuit of new materials with improved NO release function. Herein, we report the development of an NO-releasing bioerodible coating prepared from poly[bis(3-mercapto-3-methylbut-1-yl glycinyl)phosphazene] (POP-Gly-MMB), a polyphosphazene based on glycine and the naturally occurring tertiary thiol 3-mercapto-3-methylbutan-1-ol (MMB). To evaluate the NO release properties of this material, the thiolated polymer POP-Gly-MMB-SH was applied as a coating to glass substrates and subsequently converted to the NO-forming S-nitrosothiol (RSNO) derivative (POP-Gly-MMB-NO) by immersion in a mixture of tert-butyl nitrite (t-BuONO) and pentane. NO release flux from the coated substrates was determined by chemiluminescence-based NO measurement and was found to remain in a physiologically relevant range for up to 2 weeks (6.5-0.090 nmol of NO·min·cm) when immersed in pH 7.4 phosphate-buffered saline (PBS) at 37 °C. Furthermore, the coating exhibited an overall NO storage capacity of 0.89 ± 0.09 mmol·g (4.3 ± 0.6 μmol·cm). Erosion of POP-Gly-MMB-NO in PBS at 37 °C over 6 weeks results in 14% mass loss, and time-of-flight mass spectrometry (TOF-MS) was used to characterize the organic products of hydrolytic degradation as glycine, MMB, and several related esters. The comparatively long-term NO release and high storage capacity of POP-Gly-MMB-NO coatings suggest potential as a source of therapeutic NO for biomedical applications. 10.1021/acsami.6b12888
Synthesis and NMR characterization of new hyaluronan-based NO donors. Di Meo Chiara,Capitani Donatella,Mannina Luisa,Brancaleoni Enzo,Galesso Devis,De Luca Gilda,Crescenzi Vittorio Biomacromolecules Nitric oxide (NO) and hyaluronic acid (HA), two species widely different in terms of molecular complexity and biological competence, are both known to play an important role in the wound healing process. To combine the properties of HA and NO, we synthesized new NO-donors based on hyaluronic acid derivatives exhibiting a controlled NO-release under physiological conditions (in vitro tests). Since two molecules of NO can form a covalent bond with secondary amines to yield structures, named NONO-ates, able to release NO in solution, we used spermidine bound to HA as the NO-linker. The HA-spermidine derivative was obtained by controlled HA amidation in aqueous media, activating the biopolymer carboxylate groups with a water soluble carbodiimide. The resulting derivative, soluble in water, was fully characterized by high field 1H and 13C NMR spectroscopy. The amount of grafting of spermidine on HA was determined by integration of suitable 1H NMR signals. In addition, cross-linked derivatives of HA were synthesized by the Ugi's four-component reaction using formaldehyde, cyclohexylisocyanide, and spermidine. The HA-spermidine networks were characterized by 13C CP-MAS NMR spectroscopy. The degree of cross-linking of the networks was also determined. Finally, the release of NO from the swollen hydrogels freshly saturated with NO, in contact with aqueous media, was monitored by means of UV spectrophotometric measurements. 10.1021/bm050904i
Topical S-nitrosoglutathione-releasing hydrogel improves healing of rat ischaemic wounds. Georgii J L,Amadeu T P,Seabra A B,de Oliveira M G,Monte-Alto-Costa A Journal of tissue engineering and regenerative medicine Topical application of the nitric oxide (NO) donor S-nitrosoglutathione (GSNO) is known to exert beneficial effects on wound healing. The aim of this study was to evaluate, for the first time, the effect of topical application of GSNO on the healing of ischaemic wounds. Wistar rats were submitted to two parallels incisions on their backs; the skin was separated from the underlying tissue, the incisions were sutured and an excisional wound was made between the parallel incisions to create an ischaemic condition surrounding the wound. The animals were separated into a control group, which received a hydrogel vehicle without GSNO, and a GSNO-treated group, which received a GSNO-containing hydrogel. The animals were treated for 7 days consecutively with one daily application. The GSNO-treated group displayed higher rates of wound contraction and re-epithelization, lower amounts of inflammatory cells, an increase in collagen fibre density and organization and a decrease in the neovascularization compared to control. These results show that topical application of GSNO is effective in the treatment of ischaemic wounds, leading to a significant improvement in the wound healing. Therefore, topical GSNO-containing hydrogels have potential for the therapeutic treatment of ischaemic diabetic and venous ulcers. 10.1002/term.353
Nitric Oxide (NO)-Releasing Macromolecules: Rational Design and Biomedical Applications. Frontiers in chemistry Nitric oxide (NO) has been recognized as a ubiquitous gaseous transmitter and the therapeutic potential has nowadays received increasing interest. However, NO cannot be easily directly administered due to its high reactivity in air and high concentration-dependent physiological roles. As such, a plethora of NO donors have been developed that can reversibly store and release NO under specific conditions. To enhance the stability and modulate the NO release profiles, small molecule-based NO donors were covalently linked to polymeric scaffolds, rendering them with multifunctional integration, prolonged release durations, and optimized therapeutic outcomes. In this minireview, we highlight the recent achievements of NO-releasing macromolecules in terms of chemical design and biomedical applications. We hope that more efforts could be devoted to this emerging yet promising field. 10.3389/fchem.2019.00530
Progress and Promise of Nitric Oxide-Releasing Platforms. Advanced science (Weinheim, Baden-Wurttemberg, Germany) Nitric oxide (NO) is a highly potent radical with a wide spectrum of physiological activities. Depending on the concentration, it can enhance endothelial cell proliferation in a growth factor-free medium, mediate angiogenesis, accelerate wound healing, but may also lead to tumor progression or induce inflammation. Due to its multifaceted role, NO must be administered at a right dose and at the specific site. Many efforts have focused on developing NO-releasing biomaterials; however, NO short half-life in human tissues only allows this molecule to diffuse over short distances, and significant challenges remain before the full potential of NO can be realized. Here, an overview of platforms that are engineered to release NO via catalytic or noncatalytic approaches is presented, with a specific emphasis on progress reported in the past five years. A number of NO donors, natural enzymes, and enzyme mimics are highlighted, and recent promising developments of NO-releasing scaffolds, particles, and films are presented. In particular, key parameters of NO delivery are discussed: 1) NO payload, 2) maximum NO flux, 3) NO release half-life, 4) time required to reach maximum flux, and 5) duration of NO release. Advantages and drawbacks are reviewed, and possible further developments are suggested. 10.1002/advs.201701043
Nitric oxide effects on the function of aged cells ex vivo and in vivo. Reed May J,Eyman Daniel,Karres Nathan In vivo (Athens, Greece) BACKGROUND:Angiogenesis is impaired in most aged tissues. Accordingly, there is great interest in interventions that improve the ability of aged cells to undergo blood vessel formation and subsequent tissue repair. MATERIALS AND METHODS:Nitric oxide (NO), a mediator proposed to enhance angiogenesis, was administered (as the precursor SNAP, S-nitroso-N-acetylpenicillamine) to aortic ring explants from aged mice and to aged mice in two separate in vivo experiments: a PVA sponge implant model of angiogenesis and full thickness excisional dermal wounds. RESULTS:SNAP inhibited angiogenesis from the mouse aortic ring explants. However, there was a trend toward increased blood vessel formation in the sponges from the aged mice treated with SNAP. SNAP did not detectably enhance dermal wound healing or angiogenesis, but it significantly inhibited epidermal closure. CONCLUSION:These data underscore the complexity of using a single agent, even one with multiple mechanisms such as NO, to improve a clinical outcome such as angiogenesis or wound repair in aged animals.
Nitric oxide in wound-healing. Isenberg Jeff S,Ridnour Lisa A,Espey Michael Graham,Wink David A,Roberts David D Microsurgery Modulation of the complex process of wound-healing remains a surgical challenge. Little improvement beyond controlling infection, gentle tissue handling, and debridement of necrotic tissue has been had in the modern era. However, increasing appreciation of the process from a biomolecular perspective offers the potential for making significant strides in wound modulation. The bioactive molecule nitric oxide was found to have wide-ranging impact on cellular activities, including the cellular responses engendered by wound healing. Current research suggests that nitric oxide and several nitric oxide donors can exert biologic effects, although the particular net responses of cells contributing to wound repair are context-dependent. 10.1002/micr.20168
Local delivery of nitric oxide: targeted delivery of therapeutics to bone and connective tissues. Advanced drug delivery reviews Non-invasive treatment of injuries and disorders affecting bone and connective tissue remains a significant challenge facing the medical community. A treatment route that has recently been proposed is nitric oxide (NO) therapy. Nitric oxide plays several important roles in physiology with many conditions lacking adequate levels of NO. As NO is a radical, localized delivery via NO donors is essential to promoting biological activity. Herein, we review current literature related to therapeutic NO delivery in the treatment of bone, skin and tendon repair. 10.1016/j.addr.2012.03.002
Reactive Nitrogen Species Releasing Hydrogel for Enhanced Wound Healing. Zahid Alap Ali,Ahmed Rashid,Ur Rehman Syed Raza,Augustine Robin,Hasan Anwarul Annual International Conference of the IEEE Engineering in Medicine and Biology Society. IEEE Engineering in Medicine and Biology Society. Annual International Conference Poor proliferation and migration of fibroblast, keratinocyte and endothelial cells delays the wound healing in diabetic patients and results into chronicity of wounds. Slow or decreased formation of blood vessels is another issue that increases the chronicity of non-healing wounds. These chronic wounds turn into an ulcer that may lead to limb amputation. Recently, nitric oxide (NO) has emerged as a potential agent for accelerating cell migration and proliferation to enhance wound healing. It increases the expression of necessary angiogenic growth factors which stimulates the proliferation and migration of major cell types involved in wound repair. Here we report the synthesis of chitosan (CS), polyvinyl alcohol (PVA) and a NO donor S-nitroso-N-acetyl-DL-penicillamine (SNAP) to enhance the wound healing activities in chronic wounds. A three-fold increase in the proliferation of 3T3 cells was observed with NO-releasing CS-PVA hydrogels. In vitro cell migration assay demonstrated a four-fold faster migration of cells to the scratched area compared to the control group. The results depict that the use of CS-PVA hydrogel impregnated with the NO donor (SNAP) can be a promising material for promoting cell migration and subsequent accelerated healing of the chronic wounds in burns and diabetic patients. 10.1109/EMBC.2019.8856469
Hydroxocobalamins as biologically compatible donors of nitric oxide implicated in the acceleration of wound healing. Bauer J A Medical hypotheses In the late 1970s, research was unfolding that implicated nitric oxide involvement in the process of vasodilation. By 1986, research culminated in the identification of nitric oxide as the endothelium-derived relaxing factor responsible for the maintenance of vascular tone, thus implicating nitric oxide as a potential wound-healing agent. Biomedical researchers involved in wound-healing research quickly embraced the utility of developing a polymeric donor of nitric oxide which would enhance the wound-healing process. Several synthetic nitric oxide donors have been developed, dubbed 'NONOates', which have achieved great success in delivering nitric oxide to wounds. However, the impact on wound healing has been ambiguous and deemed antagonistic to the immune system in some cases. The propensity for the immune system to reject 'non-self' is a major factor in evaluating the usefulness of synthetic polymeric nitric oxide donors. The necessity of natural-product nitric oxide donors is apparent when examining the complications which are possible in a synthetic delivery system. Given the affinity nitric oxide has for transition metals, and the biological availability of transition-metal-centered products in vivo, it seems logical to pursue a transition-metal nitric oxide donor which is biologically friendly. Vitamin B12a (hydroxocobalamin), a natural product, offers an ideal environment to serve as a donor of nitric oxide.
Synthetic, Natural, and Semisynthetic Polymer Carriers for Controlled Nitric Oxide Release in Dermal Applications: A Review. Gutierrez Cisneros Carolina,Bloemen Veerle,Mignon Arn Polymers Nitric oxide (NO•) is a free radical gas, produced in the human body to regulate physiological processes, such as inflammatory and immune responses. It is required for skin health; therefore, a lack of NO• is known to cause or worsen skin conditions related to three biomedical applications- infection treatment, injury healing, and blood circulation. Therefore, research on its topical release has been increasing for the last two decades. The storage and delivery of nitric oxide in physiological conditions to compensate for its deficiency is achieved through pharmacological compounds called NO-donors. These are further incorporated into scaffolds to enhance therapeutic treatment. A wide range of polymeric scaffolds has been developed and tested for this purpose. Hence, this review aims to give a detailed overview of the natural, synthetic, and semisynthetic polymeric matrices that have been evaluated for antimicrobial, wound healing, and circulatory dermal applications. These matrices have already set a solid foundation in nitric oxide release and their future perspective is headed toward an enhanced controlled release by novel functionalized semisynthetic polymer carriers and co-delivery synergetic platforms. Finally, further clinical tests on patients with the targeted condition will hopefully enable the eventual commercialization of these systems. 10.3390/polym13050760
Supramolecular poly(acrylic acid)/F127 hydrogel with hydration-controlled nitric oxide release for enhancing wound healing. Champeau Mathilde,Póvoa Valéria,Militão Lucas,Cabrini Flávia M,Picheth Guilherme F,Meneau Florian,Jara Carlos P,de Araujo Eliana P,de Oliveira Marcelo G Acta biomaterialia 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-β, 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-β, IGF-1, SDF-1, and IL-10 gene expressions in the cicatricial tissue after topical treatment with a PAA:F127/GSNO hydrogel. 10.1016/j.actbio.2018.05.025
Functionalized GO Nanovehicles with Nitric Oxide Release and Photothermal Activity-Based Hydrogels for Bacteria-Infected Wound Healing. ACS applied materials & interfaces Bacteria-infected wounds are attracting increasing attention, as antibiotic misuse and multidrug-resistant bacteria complicate their treatment. Herein, we reported a photothermal activity-based drug consisting of β-cyclodextrin (βCD)-functionalized graphene oxide (GO) near-infrared light-responsive nanovehicles combined with the nitric oxide donor BNN6, in a methacrylate-modified gelatin (GelMA)/hyaluronic acid graft dopamine (HA-DA) hydrogel. The synergistic effects of photothermal and gas therapies are expected to improve antibacterial efficiency and reduce drug resistance. The results revealed that GelMA/HA-DA/GO-βCD-BNN6 was an ideal antibacterial material that improved collagen deposition and angiogenesis and promoted wound healing in a mouse model of full-thickness skin repair, compared to the commercially available Aquacel Ag dressing. We developed a multifunctional nanocomposite hydrogel that exhibited antibacterial and angiogenic properties, adhesiveness, and mechanical properties that enhance the regeneration of bacteria-infected wounds. 10.1021/acsami.0c04080
Nitric Oxide Produced by the Antioxidant Activity of Verapamil Improves the Acute Wound Healing Process. Han Yu Na,Lee Yoon Jae,Kim Ki Joo,Lee Su Jin,Choi Jang Youn,Moon Suk-Ho,Rhie Jong Won Tissue engineering and regenerative medicine BACKGROUND:Verapamil is used in the treatment of hypertension, angina pectoris, cardiac arrhythmia, hypertrophic scars, and keloids to block transmembrane calcium ion flux. Verapamil has antioxidant activity, which enhances the production of nitric oxide (NO). NO promotes the proliferation of fibroblasts, keratinocytes, endothelial cells, and epithelial cells during wound healing. In this study, we investigated the effect of verapamil and its antioxidant properties on the enhancement of acute wound healing via NO. METHODS:A full-thickness wound healing model was created on the rat dorsal with a silicone ring. The wound closure rate was estimated every 2 days for 14 days. A histological study was performed to evaluate wound healing. Immunofluorescence staining was analyzed for angiogenesis. The expressions of collagen type I (COL I), collagen type III (COL III), and vascular endothelial growth factor (VEGF) were assessed by Western blot. Real-time polymerase chain reaction (qRT-PCR) was performed to examine the expression of endothelial NO synthase and inducible NO synthase, which are related to antioxidant activity in the process of wound healing. RESULTS:The wound closure rate was faster in the verapamil group compared to the control and silicone groups. Histologic analysis revealed capillaries and stratum basale in the verapamil group. Immunofluorescence staining was shown vessel formation in the verapamil group. Western blot and qRT-PCR analysis revealed high expression levels of COL I, VEGF, eNOS, and FGF in the verapamil. CONCLUSION:Verapamil's antioxidant activity enhances NO production in acute wound healing. We suggest that verapamil can be used to promote acute wound healing. 10.1007/s13770-020-00308-x
Controlled nitric oxide delivery platform based on S-nitrosothiol conjugated interpolymer complexes for diabetic wound healing. Li Yan,Lee Ping I Molecular pharmaceutics Nitric oxide (NO) is known to play a critical role in enhancing wound healing as topical NO administration has demonstrated enhanced wound healing in diabetic animal models. However, this approach has been limited by the short duration of NO release, short half-life of NO, and instability of available NO donors. To overcome these deficiencies, we have developed a new NO delivery platform based on grafting S-nitrosothiols (RSNOs), derived from endogenous glutathione (GSH) or its oligomeric derivatives, phytochelatins (PCs), onto poly(vinyl methyl ether-co-maleic anhydride) (PVMMA), and their subsequent formation of interpolymer complexes with poly(vinyl pyrrolidone) (PVP). Such interpolymer complexes provide controlled release of NO for an extended duration (>10 days) and exhibit enhanced stability in the solid state over that of free RSNOs. The existence of intermolecular hydrogen bonding in such complexes and the formation of disulfide bonds following the NO release have been confirmed by FTIR and Raman. Preliminary wound healing study in a diabetic rat model demonstrates that, with a single topical application, the present controlled release NO delivery system can effectively accelerate wound closure as compared with the control (p < 0.05). The results suggest that the present NO releasing interpolymer complexes could be potentially useful for diabetic wound healing. 10.1021/mp900237f
Nitric oxide-donating materials and their potential in pharmacological applications for site-specific nitric oxide delivery. Eroy-Reveles Aura A,Mascharak Pradip K Future medicinal chemistry The discovery of various regulatory, protective and deleterious effects of nitric oxide (NO) has prompted intense research activity in the design and synthesis of NO-donating drugs and materials that can be used to modulate the effective concentrations of NO at biological targets. To date, various NO-donating compounds have been incorporated or immobilized in biocompatible polymer matrices and such materials have been used as patches, wound dressings, coatings on blood-contacting medical devices and time-release NO drugs. Recently, attention has been drawn toward light-sensitive NO donors, such as S-nitrosothiols and metal nitrosyls, which can deliver NO at selected targets under the total control of light. The pharmacological potential of such NO-donating materials including those with immobilized photoactive NO donors are reviewed in this article. 10.4155/fmc.09.111
Nitric Oxide-Releasing -Nitrosoglutathione-Conjugated Poly(Lactic--Glycolic Acid) Nanoparticles for the Treatment of MRSA-Infected Cutaneous Wounds. Lee Juho,Kwak Dongmin,Kim Hyunwoo,Kim Jihyun,Hlaing Shwe Phyu,Hasan Nurhasni,Cao Jiafu,Yoo Jin-Wook Pharmaceutics -nitrosoglutathione (GSNO) has emerged as a potent agent for the treatment of infected cutaneous wounds. However, fabrication of GSNO-containing nanoparticles has been challenging due to its high hydrophilicity and degradability. The present study aimed to fabricate nanoparticles using newly synthesized GSNO-conjugated poly(lactic--glycolic acid) (PLGA) (GSNO-PLGA; GPNPs). Since hydrophilic GSNO was covalently bound to hydrophobic PLGA, loss of GSNO during the nanoparticle fabrication process was minimized, resulting in sufficient loading efficiency (2.32% of GSNO, 0.07 μmol/mg of NO). Real-time NO release analysis revealed biphasic NO release by GPNPs, including initial burst release within 3 min and continuous controlled release for up to 11.27 h, due to the differential degradation rates of the -SNO groups located at the surface and inside of GPNPs. Since GPNPs could deliver NO more efficiently than GSNO in response to increased interaction with bacteria, the former showed enhanced antibacterial effects against methicillin-resistant (MRSA) at the same equivalent concentrations of NO. Finally, the facilitating effects of GPNPs on infected wound healing were demonstrated in MRSA-challenged full-thickness wound mouse model. Collectively, the results suggested GPNPs as an ideal nanoparticle formulation for the treatment of MRSA-infected cutaneous wounds. 10.3390/pharmaceutics12070618
The use of nitric oxide releasing nanoparticles as a treatment against Acinetobacter baumannii in wound infections. Mihu Mircea Radu,Sandkovsky Uriel,Han George,Friedman Joel M,Nosanchuk Joshua D,Martinez Luis R Virulence Acinetobacter baumannii (Ab) is a frequent cause of hospital acquired pneumonia and recently has increased in incidence as the causative agent of severe disease in troops wounded in Afghanistan and Iraq. Ab clinical isolates are frequently extremely resistant to antimicrobials, significantly complicating our capacity to treat infections due to this pathogen. Hence, the development of innovative therapeutics targeting mechanisms to which the bacteria are unlikely to evolve resistance is urgently needed. We examined the capacity of a nitric oxide-releasing nanoparticle (NO-np) to treat wounds infected with Ab. We found that the NO-nps were therapeutic in an experimental Ab murine wound model. Treatment with NO-nps significantly accelerated healing of infected wounds. Histological study demonstrated that NO-np treatment reduced suppurative inflammation, decreased microbial burden, and reduced the degradation of collagen. Furthermore, NO-np treatment alters the local cytokine milieu. In sum, we demonstrated that the NO-nps are an easily administered topical antimicrobial for the treatment of Ab wound infections, and our findings suggest that NO-nps may also be ideal for use in combat or disaster situations. 10.4161/viru.1.2.10038
Nitric oxide-containing nanoparticles as an antimicrobial agent and enhancer of wound healing. Weller Richard B The Journal of investigative dermatology Nitric oxide (NO) is known to be involved in wound healing and to have antimicrobial actions. Developing a means of delivering a diatomic, gaseous free radical has been a technical challenge. Using a combination of techniques, Martinez and colleagues have developed an ingenious method of storing NO, and they demonstrate its efficacy in treating infected wounds. 10.1038/jid.2009.149
Light-triggered nitric oxide (NO) release from photoresponsive polymersomes for corneal wound healing. Chemical science Polymersomes have been extensively used in the delivery of both small and macromolecular payloads. However, the controlled delivery of gaseous therapeutics (, nitric oxide, NO) remains a grand challenge due to its difficulty in loading of gaseous payloads into polymersomes without premature leakage. Herein, NO-releasing vesicles could be fabricated the self-assembly of NO-releasing amphiphiles, which were synthesized by the direct polymerization of photoresponsive NO monomers (abbreviated as NBN, NBN, and BN). These monomers were rationally designed through the integration of the photoresponsive behavior of -nitrosoamine moieties and the self-immolative chemistry of 4-aminobenzyl alcohol derivatives, which outperformed conventional NO donors such as diazeniumdiolates (NONOates) and -nitrosothiols (SNOs) in terms of ease of preparation, stability of storage, and controllability of NO release. The unique design made it possible to selectively release NO by a light stimulus and to regulate the NO release rates. Importantly, the photo-mediated NO release could be manipulated in living cells and showed promising applications in the treatment of corneal wounds. In addition to delivering NO, the current design enabled the synergistic delivery of NO and other therapeutic payloads by taking advantage of NO release-mediated traceless crosslinking of the vesicles. 10.1039/c9sc04039k
Effects of nitric oxide on skin burn wound healing. Zhu Haifeng,Wei Xiaofei,Bian Ka,Murad Ferid Journal of burn care & research : official publication of the American Burn Association Increasing evidence showed the important role of nitric oxide (NO) in skin repair and reconstruction. In this report, we investigated the effects of NO on 2nd degree burn wound of mice with a newly developed topical NO-gel. Using regular hematoxylin&eosin staining and immunohistochemistry, we determined the effects of NO on wound closure, hair follicle regeneration, collagen deposition, angiogenesis, and inflammatory cell infiltration in the wound of mice during wound healing. NO treatment significantly accelerated re-epithelialization by 50%, which has resulted in a markedly faster wound closure than that in control group. NO significantly promoted follicle stem cell recruitment, a key player in re-epithelialization. In addition, hair follicle regeneration also was enhanced by NO treatment in mice. As we have reported with rat model, NO treatment significantly increased the number of procollagen-expressing fibroblasts, which peaked by day 10 after burn wound. We also demonstrated an increase of angiogenesis in NO treated wounds compared with that in the control group during wound healing. Finally, we found that the NO gel promoted wound bed infiltration and retention of inflammatory cells that are a major source of growth factors and cytokines during the healing processes. These observations suggest that NO released from a topical preparation has the potential to enhance burn wound healing by regulation of many cellular processes in the skin. 10.1097/BCR.0b013e3181848119
The purview of nitric oxide nanoparticle therapy in infection and wound healing. Sanchez David Alejandro,Nosanchuk Joshua,Friedman Adam Nanomedicine (London, England) 10.2217/nnm.12.67
Oral or topical administration of L-arginine changes the expression of TGF and iNOS and results in early wounds healing. Jerônimo Márcio Sousa,Barros Adria do Prado,MoritaI Vinícius Elord Zen,Alves Erika Oliveira,Souza Nathalia Lobão Barroso de,Almeida Rosane Mansan de,Nóbrega Yanna Karla Medeiros,Cavalcanti Florêncio Figueiredo,Amorin Rivadávio,Borin Maria de Fátima,Bocca Anamélia Lorenzetti Acta cirurgica brasileira PURPOSE::To evaluate the contribution of L-arginine oral or topical rout of administration in the surgical wound healing process. METHODS::L-arginine was orally or topically administrated to mice after a laparotomy model procedure. The wounds were analyzed to evaluate the granulation tissue by HE analysis, collagen deposition, iNOS and cytokines production by immunochemisyry on wound progress. Mice used in this model were healthy, immunosupressed or diabetic and all of them were treated with different concentration of L-arginine and rout of administration. RESULTS::Suggested that groups treated with L-arginine orally or topically improved wound repair when compared with non-treatad mice. L- arginine treatment stimulated TGF-β and restricted NO production leading to a mild Th1 response and collagen deposition in injured area, when it was orally administrated. Topical administration decreased IL-8 and CCR1 expression by wound cells but did not interfere with TNF-α and IL-10 production, ratifying the decrease of inflammatory response, the oral administration however, presented a higher iNOS and TGF-β expression then. L-arginine treatment also improved the improved the wound healing in immunosupressed or diabetic mice. CONCLUSION::L-arginine administrated orally or topically can be considered an important factor in the recuperation of tissues. 10.1590/S0102-865020160090000003
The effect of nitric oxide releasing cream on healing pressure ulcers. Iranian journal of nursing and midwifery research BACKGROUND:Pressure ulcer is one of the main concerns of nurses in medical centers around the world, which, if untreated, causes irreparable problems for patients. In recent years, nitric oxide (NO) has been proposed as an effective method for wound healing. This study was conducted to determine the effect of nitric oxide on pressure ulcer healing. MATERIALS AND METHODS:In this clinical trial, 58 patients with pressure ulcer at hospitals affiliated to Ahvaz Jundishapur University of Medical Sciences were homogenized and later divided randomly into two groups of treatment (nitric oxide cream; n = 29) and control (placebo cream; n = 29). In this research, the data collection tool was the Pressure Ulcer Scale for Healing (PUSH). At the outset of the study (before using the cream), the patients' ulcers were examined weekly in terms of size, amount of exudates, and tissue type using the PUSH tool for 3 weeks. By integrating these three factors, wound healing was determined. Data were analyzed using SPSS. RESULTS:Although no significant difference was found in terms of the mean of score size, the amount of exudates, and the tissue type between the two groups, the mean of total score (healing) between the two groups was statistically significant (P = 0.04). CONCLUSIONS:Nitric oxide cream seems to accelerate wound healing. Therefore, considering its easy availability and cost-effectiveness, it can be used for treating pressure ulcers in the future. 10.4103/1735-9066.180389
Nanomaterial Nitric Oxide Delivery in Traumatic Orthopedic Regenerative Medicine. Anastasio Albert Thomas,Paniagua Ariana,Diamond Carrie,Ferlauto Harrison R,Fernandez-Moure Joseph S Frontiers in bioengineering and biotechnology Achieving bone fracture union after trauma represents a major challenge for the orthopedic surgeon. Fracture non-healing has a multifactorial etiology and there are many risk factors for non-fusion. Environmental factors such as wound contamination, infection, and open fractures can contribute to non-healing, as can patient specific factors such as poor vascular status and improper immunologic response to fracture. Nitric oxide (NO) is a small, neutral, hydrophobic, highly reactive free radical that can diffuse across local cell membranes and exert paracrine functions in the vascular wall. This molecule plays a role in many biologic pathways, and participates in wound healing through decontamination, mediating inflammation, angiogenesis, and tissue remodeling. Additionally, NO is thought to play a role in fighting wound infection by mitigating growth of both Gram negative and Gram positive pathogens. Herein, we discuss recent developments in NO delivery mechanisms and potential implications for patients with bone fractures. NO donors are functional groups that store and release NO, independent of the enzymatic actions of NOS. Donor molecules include organic nitrates/nitrites, metal-NO complexes, and low molecular weight NO donors such as NONOates. Numerous advancements have also been made in developing mechanisms for localized nanomaterial delivery of nitric oxide to bone. NO-releasing aerogels, sol- gel derived nanomaterials, dendrimers, NO-releasing micelles, and core cross linked star (CCS) polymers are all discussed as potential avenues of NO delivery to bone. As a further target for improved fracture healing, 3d bone scaffolds have been developed to include potential for nanoparticulated NO release. These advancements are discussed in detail, and their potential therapeutic advantages are explored. This review aims to provide valuable insight for translational researchers who wish to improve the armamentarium of the feature trauma surgeon through use of NO mediated augmentation of bone healing. 10.3389/fbioe.2020.592008
Enzyme-controllable delivery of nitric oxide from a molecular hydrogel. Gao Jie,Zheng Wenting,Zhang Jimin,Guan Di,Yang Zhimou,Kong Deling,Zhao Qiang Chemical communications (Cambridge, England) A β-galactosidase-responsive molecular hydrogelator of a nitric oxide (NO) donor can release NO in a controllable manner to improve wound healing. 10.1039/c3cc45666h
Nitric oxide in myogenesis and therapeutic muscle repair. De Palma Clara,Clementi Emilio Molecular neurobiology Nitric oxide is a short-lived intracellular and intercellular messenger. The first realisation that nitric oxide is important in physiology occurred in 1987 when its identity with the endothelium-derived relaxing factor was discovered. Subsequent studies have shown that nitric oxide possesses a number of physiological functions that are essential not only to vascular homeostasis but also to neurotransmission, such as in the processes of learning and memory and endocrine gland regulation, as well as inflammation and immune responses. The discovery in 1995 that a splice variant of the neuronal nitric oxide synthase is localised at the sarcolemma via the dystrophin-glycoprotein complex and of its displacement in Duchenne muscular dystrophy has stimulated a host of studies exploring the role of nitric oxide in skeletal muscle physiology. Recently, nitric oxide has emerged as a relevant messenger also of myogenesis that it regulates at several key steps, especially when the process is stimulated for muscle repair following acute and chronic muscle injuries. Here, we will review briefly the mechanisms and functions of nitric oxide in skeletal muscle and discuss its role in myogenesis, with specific attention to the promising nitric oxide-based approaches now being explored at the pre-clinical and clinical level for the therapy of muscular dystrophy. 10.1007/s12035-012-8311-8
Nitric oxide: a newly discovered function on wound healing. Luo Jian-dong,Chen Alex F Acta pharmacologica Sinica Wound healing impairment represents a particularly challenging clinical problem to which no efficacious treatment regimens currently exist. The factors ensuring appropriate intercellular communication during wound repair are not completely understood. Although protein-type mediators are well-established players in this process, emerging evidence from both animal and human studies indicates that nitric oxide (NO) plays a key role in wound repair. The beneficial effects of NO on wound repair may be attributed to its functional influences on angiogenesis, inflammation, cell proliferation, matrix deposition, and remodeling. Recent findings from in vitro and in vivo studies of NO on wound repair are summarized in this review. The unveiled novel mechanisms support the use of NO-containing agents and/or NO synthase gene therapy as new therapeutic regimens for impaired wound healing. 10.1111/j.1745-7254.2005.00058.x
[Advance of researches in nitric oxide biological function on wound repair]. Tang Xinyan,Yang Li,Sung K L Paul Sheng wu yi xue gong cheng xue za zhi = Journal of biomedical engineering = Shengwu yixue gongchengxue zazhi Nitric oxide (NO) is a short-life free radical that acts as the small biological molecule, and exists in body extensively. Since its discovery over 20 years ago, NO has been found to play an important regulation role in angiogenesis, nerve and immune system. The subsequent studies also showed that NO exerted an important biological action in wound repairing and healing, which involved in the following phases of wound repair, inflammation, cell proliferation, matrix deposition and remodeling. This paper reviews recent findings from in vitro & in vivo studies of NO in wound repair, and the biological function and mechanisms of NO in wound repair.
S-Nitrosothiols as Platforms for Topical Nitric Oxide Delivery. Ganzarolli de Oliveira Marcelo Basic & clinical pharmacology & toxicology Nitric oxide (NO) is a small radical species involved in several fundamental physiological processes, including the control of vascular tone, the immune response and neuronal signalling. Endothelial dysfunction with the decreased NO bioavailability is the underlying cause of several diseases and has led to the development of a wide range of systemic NO donor compounds to lower the blood pressure and control hypertensive crises. However, several potential therapeutic actions of NO, not related to the cardiovascular system, demand exclusively local actions. Primary S-nitrosothiols (RSNOs) are endogenously found NO carriers and donors and have emerged as platforms for the localized delivery of NO in topical applications. Formulations for this purpose have evolved from low molecular weight RSNOs incorporated in polymeric films, hydrogels and viscous vehicles, to polymeric RSNOs where the SNO moiety is covalently bound to the polymer backbone. The biological actions displayed by these formulations include the increase in dermal vasodilation, the acceleration of wound healing, the killing of infectious microorganisms and an analgesic action against inflammatory pain. This MiniReview focuses on the state of the art of experimental topical formulations for NO delivery based on S-nitrosothiols and their potential therapeutic applications. 10.1111/bcpt.12588
Nitric Oxide-Releasing Hyaluronic Acid as an Antibacterial Agent for Wound Therapy. Maloney Sara E,McGrath Kyle V,Ahonen Mona Jasmine R,Soliman Daniel S,Feura Evan S,Hall Hannah R,Wallet Shannon M,Maile Robert,Schoenfisch Mark H Biomacromolecules Taking advantage of their respective wound-healing roles in physiology, the dual activity of hyaluronic acid (HA) and nitric oxide (NO) was combined to create a single-agent wound therapeutic. Carboxylic acid groups of HA (6 and 90 kDa) were chemically modified with a series of alkylamines via carbodiimide chemistry to provide secondary amines for subsequent -diazeniumdiolate NO donor formation. The resulting NO-releasing HA derivatives stored 0.3-0.6 μmol NO mg and displayed diverse release kinetics (5-75 min NO-release half-lives) under physiological conditions. The 6 kDa HA with terminal primary amines and intermediate release kinetics exhibited broad-spectrum bactericidal activity against common wound pathogens, including planktonic methicillin-resistant as well as planktonic and biofilm-based multidrug-resistant . The treatment of infected murine wounds with NO-releasing HA facilitated more rapid wound closure and decreased the quantity of the genetic material in the remaining wound tissue. Hyaluronidase readily degraded the HA derivatives, indicating that NO donor modification did not prohibit endogenous biodegradation pathways. 10.1021/acs.biomac.0c01583
Nitrosyl-cobinamide (NO-Cbi), a new nitric oxide donor, improves wound healing through cGMP/cGMP-dependent protein kinase. Spitler Ryan,Schwappacher Raphaela,Wu Tao,Kong Xiangduo,Yokomori Kyoko,Pilz Renate B,Boss Gerry R,Berns Michael W Cellular signalling Nitric oxide (NO) donors have been shown to improve wound healing, but the mechanism is not well defined. Here we show that the novel NO donor nitrosyl-cobinamide (NO-Cbi) improved in vitro wound healing in several cell types, including an established line of lung epithelial cells and primary human lung fibroblasts. On a molar basis, NO-Cbi was more effective than two other NO donors, with the effective NO-Cbi concentration ranging from 3 to 10μM, depending on the cell type. Improved wound healing was secondary to increased cell migration and not cell proliferation. The wound healing effect of NO-Cbi was mediated by cGMP, mainly through cGMP-dependent protein kinase type I (PKGI), as determined using pharmacological inhibitors and activators, and siRNAs targeting PKG type I and II. Moreover, we found that Src and ERK were two downstream mediators of NO-Cbi's effect. We conclude that NO-Cbi is a potent inducer of cell migration and wound closure, acting via cGMP, PKG, Src, and extracellular signal regulated kinase (ERK). 10.1016/j.cellsig.2013.07.029
Nitric oxide-releasing nanoparticles accelerate wound healing in NOD-SCID mice. Blecher Karin,Martinez Luis R,Tuckman-Vernon Chaim,Nacharaju Parimala,Schairer David,Chouake Jason,Friedman Joel M,Alfieri Alan,Guha Chandan,Nosanchuk Joshua D,Friedman Adam J Nanomedicine : nanotechnology, biology, and medicine 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. 10.1016/j.nano.2012.02.014
Hydrogel cross-linking-programmed release of nitric oxide regulates source-dependent angiogenic behaviors of human mesenchymal stem cell. Kang Mi-Lan,Kim Hye-Seon,You Jin,Choi Young Sik,Kwon Byeong-Ju,Park Chan Hee,Baek Wooyeol,Kim Min Sup,Lee Yong Jae,Im Gun-Il,Yoon Jeong-Kee,Lee Jung Bok,Sung Hak-Joon Science advances Angiogenesis is stimulated by nitric oxide (NO) production in endothelial cells (ECs). Although proangiogenic actions of human mesenchymal stem cells (hMSCs) have been extensively studied, the mechanistic role of NO in this action remains obscure. Here, we used a gelatin hydrogel that releases NO upon crosslinking by a transglutaminase reaction ("NO gel"). Then, the source-specific behaviors of bone marrow versus adipose tissue-derived hMSCs (BMSCs versus ADSCs) were monitored in the NO gels. NO inhibition resulted in significant decreases in their angiogenic activities. The NO gel induced pericyte-like characteristics in BMSCs in contrast to EC differentiation in ADSCs, as evidenced by tube stabilization versus tube formation, 3D colocalization versus 2D coformation with EC tube networks, pericyte-like wound healing versus EC-like vasculogenesis in gel plugs, and pericyte versus EC marker production. These results provide previously unidentified insights into the effects of NO in regulating hMSC source-specific angiogenic mechanisms and their therapeutic applications. 10.1126/sciadv.aay5413
Nitric Oxide-Releasing Biomaterials for Biomedical Applications. Zhou Xin,Zhang Jimin,Feng Guowei,Shen Jie,Kong Deling,Zhao Qiang Current medicinal chemistry Nitric oxide (NO), as an essential signaling molecule, participates in various physiological processes such as cardiovascular homeostasis, neuronal transmission, immunomodulation, and tumor growth. The multiple role of NO in physiology and pathophysiology has triggered a massive interest in the strategies of delivering exogenous NO for biomedical applications. Hence, different kinds of NO prodrugs have been developed up to date, including diazeniumdiolates, S-nitrosothiol, metal-nitrosyl, nitrobenzene, and so on. However, the clinical application of these low molecular weight NO donors has been restricted due to the problems of burst release, low payloads, and untargeted delivery. The delivery of NO by biomaterialbased carrier offers a beneficial strategy to realize the controlled and sustained delivery of NO to the targeted tissues or organs. In detail, NO-donor prodrugs have been attached and loaded to diverse biomaterials to fabricate nanoparticles, hydrogels, and coating platforms by means of physical, chemical, or supramolecular techniques. These NO-releasing biomaterials hold promise for a number of biomedical applications ranging from therapy of the ischemic disease and several types of cancer to cardiovascular devices and wound dressing. First, surface coating with NO-releasing biomaterials could mimic the physiological function of vascular endothelium, therefore promoting vascularization and improving the patency of cardiovascular implants. Next, because NO also mediates many important processes that take place after cutaneous injury, NO-releasing biomaterials could serve as ideal wound dressing to accelerate tissue regeneration. Finally, biomaterials enable localized delivery of high dose of NO to tumors in a sustained manner, thus generating potent tumoricidal effect. In this review, we will summarize the progress of different NO-releasing biomaterials, and highlight their biomedical applications with a hope to inspire new perspectives in the area of biomaterial-based NO-delivery systems.
Biotemplated Synthesis and Characterization of Mesoporous Nitric Oxide-Releasing Diatomaceous Earth Silica Particles. Grommersch Bryan M,Pant Jitendra,Hopkins Sean P,Goudie Marcus J,Handa Hitesh ACS applied materials & interfaces Diatomaceous earth (DE), a nanoporous silica material composed of fossilized unicellular marine algae, possesses unique mechanical, molecular transport, optical, and photonic properties exploited across an array of biomedical applications. The utility of DE in these applications stands to be enhanced through the incorporation of nitric oxide (NO) technology shown to modulate essential physiological processes. In this work, the preparation and characterization of a biotemplated diatomaceous earth-based nitric oxide delivery scaffold are described for the first time. Three aminosilanes [(3-aminopropyl)triethoxysilane (APTES), N-(6-aminohexyl)aminomethyltriethoxysilane (AHAMTES), and 3-aminopropyldimethylethoxysilane (APDMES)] were evaluated for their ability to maximize NO loading via the covalent attachment of N-acetyl-d-penicillamine (NAP) to diatomaceous earth. The use of APTES cross-linker resulted in maximal NAP tethering to the DE surface, and NAP-DE was converted to NO-releasing S-nitroso-N-acetyl-penicillamine (SNAP)-DE by nitrosation. The total NO loading of SNAP-DE was determined by chemiluminescence to be 0.0372 ± 0.00791 μmol/mg. Retention of diatomaceous earth's unique mesoporous morphology throughout the derivatization was confirmed by scanning electron microscopy. SNAP-DE exhibited 92.95% killing efficiency against Gram-positive bacteria Staphylococcus aureus as compared to the control. The WST-8-based cytotoxicity testing showed no negative impact on mouse fibroblast cells, demonstrating the biocompatible potential of SNAP-DE. The development of NO releasing diatomaceous earth presents a unique means of delivering tunable levels of NO to materials across the fields of polymer chemistry, tissue engineering, drug delivery, and wound healing. 10.1021/acsami.7b15967
Delivering nitric oxide with nanoparticles. Quinn John F,Whittaker Michael R,Davis Thomas P Journal of controlled release : official journal of the Controlled Release Society While best known for its important signalling functions in human physiology, nitric oxide is also of considerable therapeutic interest. As such, nanoparticle-based systems which enable the sustained exogenous delivery of nitric oxide have been the subject of considerable investigation in recent years. Herein we review the various nanoparticle systems that have been used to date for nitric oxide delivery, and explore the array of potential therapeutic applications that have been reported. Specifically, we discuss the modification of sol-gel based silica particles, functionalised metal/metal oxide nanoparticles, polymer-coated metal nanoparticles, dendrimers, micelles and star polymers to impart nitric oxide release capability. We also consider the various areas in which therapeutic applications are envisaged: wound healing, antimicrobial applications, cardiovascular treatments, sexual medicine and cancer treatment. Finally, we discuss possible future directions for this versatile and potentially important technology. 10.1016/j.jconrel.2015.02.007
In Situ Hydrogel-Forming/Nitric Oxide-Releasing Wound Dressing for Enhanced Antibacterial Activity and Healing in Mice with Infected Wounds. Lee Juho,Hlaing Shwe Phyu,Cao Jiafu,Hasan Nurhasni,Ahn Hye-Jin,Song Ki-Won,Yoo Jin-Wook Pharmaceutics The eradication of bacteria from wound sites and promotion of healing are essential for treating infected wounds. Nitric oxide (NO) is desirable for these purposes due to its ability to accelerate wound healing and its broad-spectrum antibacterial effects. We developed an in situ hydrogel-forming/NO-releasing powder dressing (NO/GP), which is a powder during storage and forms a hydrogel when applied to wounds, as a novel NO-releasing formulation to treat infected wounds. An NO/GP fine powder (51.5 μm) was fabricated by blending and micronizing S-nitrosoglutathione (GSNO), alginate, pectin, and polyethylene glycol (PEG). NO/GP remained stable for more than four months when stored at 4 or 37 °C. When applied to wounds, NO/GP absorbed wound fluid and immediately converted to a hydrogel. Additionally, wound fluid triggered a NO release from NO/GP for more than 18 h. The rheological properties of hydrogel-transformed NO/GP indicated that NO/GP possesses similar adhesive properties to marketed products (Vaseline). NO/GP resulted in a 6-log reduction in colony forming units (CFUs) of methicillin resistant (MRSA) and which are representative drug-resistant gram-positive and -negative bacteria, respectively. The promotion of wound healing by NO/GP was demonstrated in mice with full-thickness wounds challenged with MRSA and . Thus, NO/GP is a promising formulation for the treatment of infected wounds. 10.3390/pharmaceutics11100496
Nitric oxide coating polypropylene mesh increases angiogenesis and reduces inflammatory response and apoptosis. Prudente Alessandro,Favaro Wágner José,Reis Leonardo Oliveira,Riccetto Cássio Luis Zanettini International urology and nephrology OBJECTIVES:To evaluate the effect of implanted S-nitrosoglutathione (GSNO) coating polypropylene mesh in foreign-body response of rats. METHODS:Thirty female rats underwent to subcutaneous implant of five polypropylene (PP) fragments: uncoated PP (control); PP polyvinylalcohol (PVA) coated and PP PVA + GSNO (1, 10 and 70 mMol) coated. After euthanasia (4 and 30 days), eight slides were prepared from each animal: hematoxylin-eosin (inflammatory response); unstained (birefringence collagen evaluation); TUNEL technique (apoptosis); and five for immunohistochemical processing: CD-31 (angiogenesis), IL-1 and TNF-α (proinflammatory cytokynes), iNOS (NO synthesis) and MMP-2 (collagen metabolism). The inflammation area, birefringence index, apoptotic index, immunoreactivity and vessel density were objectively measured. RESULTS:Inflammatory reaction area at 4 days was 11.3, 15.2, 25.1, 17.1 and 19.3% of pure PP, PVA, GSNO 1, 10 and 70 mM, respectively, p = 0.0006 (PP × Others). At 30 days lower inflammatory area was observed in GSNO 10 and 70 mM compared to pure PP (5.3, 5.2 and 11.1%, respectively, p = 0.0001). Vessel density was higher for GSNO 1 mM (25.5%) compared to pure PP (19.6%) at 30 days only, p = 0.0081. Apoptotic index at 4 days was lower for GSNO 1 mM (49.3%) than pure PVA (60.6%), p = 0.0124. GSNO 10 and 70 mM reduced their apoptotic index at 30 days compared to 4 days (49.9 vs. 36.9 and 59.1 vs. 47.5%, respectively, p = 0.0397). Birefringence index, IL-1, TNF, MMP-2 and iNOS were not different. CONCLUSIONS:Depending on concentrations, GSNO can increase angiogenesis, reduce inflammation and apoptosis compared to pure PP, without impact on cytokine, collagen organization/metabolism and endogenous NO synthesis. 10.1007/s11255-017-1520-3
Cellulose-based biomaterials integrated with copper-cystine hybrid structures as catalysts for nitric oxide generation. Darder Margarita,Karan Anik,Real Gustavo Del,DeCoster Mark A Materials science & engineering. C, Materials for biological applications Bionanocomposite materials were developed from the assembly of polymer-coated copper-cystine high-aspect ratio structures (CuHARS) and cellulose fibers. The coating of the metal-organic materials with polyallylamine hydrochloride (PAH) allows their covalent linkage to TEMPO-oxidized cellulose by means of EDC/NHS. The resulting materials can be processed as films or macroporous foams by solvent casting and lyophilization, respectively. The films show good mechanical behavior with Young's moduli around 1.5 GPa as well as resistance in water, while the obtained foams show an open network of interconnected macropores with average diameters around 130 μm, depending on the concentration of the initial suspension, and compression modulus values around 450 kPa, similar to other reported freeze-dried nanocellulose-based aerogels. Based on these characteristics, the cellulose/PAH-CuHARS composites are promising for potential biomedical applications as implants or wound dressing materials. They have proved to be effective in the decomposition of low molecular weight S-nitrosothiols (RSNOs), similar to those existing in blood, releasing nitric oxide (NO). This effect is attributed to the presence of copper in the crystalline structure of the CuHARS building unit, which can be gradually released in the presence of redox species like ascorbic acid, typically found in blood. The resulting biomaterials can offer the interesting properties associated with NO, like antimicrobial activity as preliminary tests showed here with Escherichia coli and Staphylococcus epidermidis. In the presence of physiological concentration of RSNOs the amount of generated NO (around 360 nM) is not enough to show bactericidal effect on the studied bacteria, but it could provide other properties inherent to NO even at low concentration in the nM range like anti-inflammatory and anti-thrombotic effects. The cytotoxic effect recorded of the films on rat brain endothelial cells (BMVECs) is least significant and proves them to be friendly enough for further biological studies. 10.1016/j.msec.2019.110369
The anabolic effect of arginine on proteins in skin wound and muscle is independent of nitric oxide production. Zhang Xiao-jun,Chinkes David L,Wolfe Robert R Clinical nutrition (Edinburgh, Scotland) BACKGROUND & AIMS:L-arginine may play a role in regulation of protein metabolism. The aim of the present study was to investigate the effect of L-arginine supplementation on protein metabolism in skin wound and muscle in anesthetized rabbits. METHODS:L-[ring-(13)C(6)]phenylalanine was infused as a tracer on day 7 after ear injury, and the scalded ear and uninjured hindlimb were used as arteriovenous units to reflect protein kinetics in these two tissues. In study 1, an amino acid mixture (10% Travasol) was infused either alone at 1.5 ml/kg per h or at 0.75 ml/kg per h with supplemental L-arginine to deliver comparable amount of amino acid nitrogen. In study 2, N(omega)-nitro-L-arginine methyl ester was infused to inhibit nitric oxide synthase during the stable isotope infusion. RESULTS:In study 1, arginine supplementation increased (p<0.05) net protein balance in skin wound and muscle from -6.7+/-6.2 to -0.8+/-3.8 and from -4.4+/-2.4 to -1.9+/-1.5 micromol phenylalanine/100 g per h, respectively, indicating an anabolic effect. In study 2, the N(omega)-nitro-L-arginine methyl ester infusion markedly reduced the blood flow rate in the scalded ear and increased (p<0.05) net protein balance in skin wound and in muscle from -8.6+/-3.4 to -1.0+/-5.7 and from -3.9+/-1.3 to -2.2+/-0.5 micromol phenylalanine/100 g per h, respectively. CONCLUSIONS:Arginine supplementation increased net protein balance in skin wound and muscle by a mechanism which was independent of nitric oxide production. 10.1016/j.clnu.2008.01.006
Wound healing action of nitric oxide-releasing self-expandable collagen sponge. Póvoa Valéria C O,Dos Santos Giovanna J V P,Picheth Guilherme F,Jara Carlos P,da Silva Laura C E,de Araújo Eliana P,de Oliveira Marcelo G Journal of tissue engineering and regenerative medicine 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-β), 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. 10.1002/term.3046
Nitric Oxide Release from Antimicrobial Peptide Hydrogels for Wound Healing. Durão Joana,Vale Nuno,Gomes Salomé,Gomes Paula,Barrias Cristina C,Gales Luís Biomolecules Nitric oxide (NO) is an endogenously produced molecule that has been implicated in several wound healing mechanisms. Its topical delivery may improve healing in acute or chronic wounds. In this study an antimicrobial peptide was synthesized which self-assembled upon a pH shift, forming a hydrogel. The peptide was chemically functionalized to incorporate a NO-donor moiety on lysine residues. The extent of the reaction was measured by ninhydrin assay and the NO release rate was quantified via the Griess reaction method. The resulting compound was evaluated for its antimicrobial activity against , and its effect on collagen production by fibroblasts was assessed. Time-kill curves point to an initial increase in bactericidal activity of the functionalized peptide, and collagen production by human dermal fibroblasts when incubated with the NO-functionalized peptide showed a dose-dependent increase in the presence of the NO donor within a range of 0⁻20 μM. 10.3390/biom9010004
Nitric oxide-releasing injectable hydrogels with high antibacterial activity through in situ formation of peroxynitrite. Hoang Thi Thai Thanh,Lee Yunki,Le Thi Phuong,Park Ki Dong Acta biomaterialia Nitric oxide (NO) is an endogenous molecule with many critical biological functions that depend on its concentration. At high levels, NO provides broad-spectrum antibacterial effects through both its pathogen inhibition and killing abilities. However, its short half-life has been a great challenge to its clinical application in pharmaceutical forms. In this study, we incorporated the NO donor S-nitrosothiolated gelatin (GelSNO) into injectable gelatin-based hydrogels (GHs) to controllably release NO. Under catalysis by horseradish peroxidase, HO oxidizes phenol moieties functionalized on gelatin to quickly form phenol-phenol crosslinks that encapsulate GelSNO. Through thermal, visible light, and oxidizing agent-driven mechanisms, NO is released from the GH/GelSNO hydrogels. By varying the GelSNO concentration, the release of NO was controllable in a wide range, 0.054-2.050 μmol/mL, for up to 14 days. In addition, NO release was fine-tunable as a function of HO concentration. Notably, the in situ formation of peroxynitrite (ONOO) that produces potent antibacterial effects originated from HO residues and nitrous acid formed by NO and oxygen in aqueous solution. The Kirby-Bauer method indicated that there was an inhibition zone against both Escherichia coli and Staphylococcus aureus incubated with GH/GelSNO hydrogels. The AlarmaBlue assay showed that E. coli and S. aureus were completely killed at NO concentrations of 0.39 and 0.58 μmol/mL. Cytotoxicity tests of GH/GelSNO hydrogels on human dermal fibroblasts at the indicated bactericidal NO concentrations induced no cell toxicity. In summary, GH/GelSNO hydrogels may provide a new platform for topical delivery of NO in treating wound infections and for various biomedical applications. STATEMENT OF SIGNIFICANCE:NO is an effective antibacterial agent even in cases of antibiotic-resistant bacteria. Moreover, its intermediate, peroxynitrite, has been reported to have a much higher ability to kill bacteria. In this study, we utilized injectable GH/GelSNO hydrogels formed by HRP/HO reaction not only to control NO release but also to generate peroxynitrite in situ from released NO and HO residues. The GH/GelSNO hydrogels showed significant antibacterial ability on both gram-positive and negative bacteria, while no cytotoxicity was induced on human dermal fibroblasts. In addition, their tunable chemico-physical properties and controllable NO release within a wide range but narrow scale will make the hydrogels useful in various biomedical applications. 10.1016/j.actbio.2017.12.005
Optimized polymeric film-based nitric oxide delivery inhibits bacterial growth in a mouse burn wound model. Brisbois Elizabeth J,Bayliss Jill,Wu Jianfeng,Major Terry C,Xi Chuanwu,Wang Stewart C,Bartlett Robert H,Handa Hitesh,Meyerhoff Mark E Acta biomaterialia Nitric oxide (NO) has many biological roles (e.g. antimicrobial agent, promoter of angiogenesis, prevention of platelet activation) that make NO releasing materials desirable for a variety of biomedical applications. Localized NO release can be achieved from biomedical grade polymers doped with diazeniumdiolated dibutylhexanediamine (DBHD/N2O2) and poly(lactic-co-glycolic acid) (PLGA). In this study, the optimization of this chemistry to create film/patches that can be used to decrease microbial infection at wound sites is examined. Two polyurethanes with different water uptakes (Tecoflex SG-80A (6.2±0.7wt.%) and Tecophilic SP-60D-20 (22.5±1.1wt.%)) were doped with 25wt.% DBHD/N2O2 and 10wt.% of PLGA with various hydrolysis rates. Films prepared with the polymer that has the higher water uptake (SP-60D-20) were found to have higher NO release and for a longer duration than the polyurethane with the lower water uptake (SG-80A). The more hydrophilic polymer enhances the hydrolysis rate of the PLGA additive, thereby providing a more acidic environment that increases the rate of NO release from the NO donor. The optimal NO releasing and control SG-80A patches were then applied to scald burn wounds that were infected with Acinetobacter baumannii. The NO released from these patches applied to the wounds is shown to significantly reduce the A. baumannii infection after 24h (∼4 log reduction). The NO release patches are also able to reduce the level of transforming growth factor-β in comparison to controls, which can enhance re-epithelialization, decrease scarring and reduce migration of bacteria. The combined DBHD/N2O2 and PLGA-doped polymer patches, which could be replaced periodically throughout the wound healing process, demonstrate the potential to reduce risk of bacterial infection and promote the overall wound healing process. 10.1016/j.actbio.2014.06.032
Nitric oxide releasing chitosan-poly (vinyl alcohol) hydrogel promotes angiogenesis in chick embryo model. Zahid Alap Ali,Ahmed Rashid,Raza Ur Rehman Syed,Augustine Robin,Tariq Muhammad,Hasan Anwarul International journal of biological macromolecules The lack of angiogenic activity is one of the serious complications of chronic wounds associated with delayed wound closure, chronic ulceration, and subsequent limb amputation. Multiple lines of evidence suggest that nitric oxide (NO) produced endogenously by nitric oxide synthase pathway plays a significant role in angiogenic activity and accelerates wounds closure. In this work, chitosan (CS), polyvinyl alcohol (PVA) and S-nitroso-N-acetyl-DL-penicillamine (SNAP) hydrogel was fabricated to accelerate angiogenesis and promote healing in chronic wounds due to better wound closure potential of CS-PVA hydrogel and angiogenic properties of SNAP. The developed CS-PVA hydrogels loaded with SNAP produced a continuous and sustained supply of NO. 3T3 and HaCaT cells showed a significant increase in cell proliferation with 5‰ SNAP loaded CS-PVA hydrogel compared to the control group. Wound scratch assay resulted in four-fold faster recovery of the scratched wound area and an enhanced degree of angiogenic activity was observed in the chick embryo model with the SNAP incorporated CS-PVA hydrogel compared to the control group. The results depict that the use of CS-PVA hydrogel impregnated with SNAP could be a promising material for promoting angiogenesis followed by accelerated healing of the chronic wounds in burns and diabetic patients. 10.1016/j.ijbiomac.2019.06.136
Nitric oxide-releasing polyacrylonitrile disperses biofilms formed by wound-relevant pathogenic bacteria. Craven M,Kasper S H,Canfield M J,Diaz-Morales R R,Hrabie J A,Cady N C,Strickland A D Journal of applied microbiology AIMS:To test the antimicrobial and antibiofilm properties of a nitric oxide (NO)-releasing polymer against wound-relevant bacterial pathogens. METHODS AND RESULTS:Using a variety of 96-well plate assay systems that include standard well plates and the minimum biofilm eradication concentration biofilm assay well plate, a NO-releasing polymer based on (poly)acrylonitrile (PAN/NO) was studied for antimicrobial and antibiofilm activity against the common wound pathogens Pseudomonas aeruginosa (PAO1), Staphylococcus aureus (Mu50) and Enterococcus faecalis (V583). The polymer was capable of dispersing single-species biofilms of Ps. aeruginosa as well as a more clinically relevant multispecies biofilm that incorporates Ps. aeruginosa along with Staph. aureus and Ent. faecalis. PAN/NO also synergistically enhanced the susceptibility of the multispecies biofilms to the common broad-spectrum antibiotic, ciprofloxacin. Multiple in vitro biocompatibility assays show that PAN/NO has limited potential for mammalian cytotoxicity. CONCLUSION:This study demonstrates the feasibility of utilizing the NO-releasing polymer, PAN/NO, to manage biofilms formed by wound-relevant pathogens, and provides proof-of-concept for use of this NO-releasing polymer platform across multiple disciplines where bacterial biofilms pose significant problems. SIGNIFICANCE AND IMPACT OF STUDY:In the clinical sector, bacterial biofilms represent a substantial treatment challenge for health care professionals and are widely recognized as a key factor in prolonging patient morbidity. This study highlights the potential role for the ubiquitous signalling molecule nitric oxide (NO) as an antibiofilm therapy. 10.1111/jam.13059
S-nitrosothiol-terminated poly(vinyl alcohol): Nitric oxide release and skin blood flow response. Giglio Leonardo P,Picheth Guilherme F,Løvschall Kaja Borup,Zelikin Alexander N,de Oliveira Marcelo G Nitric oxide : biology and chemistry Polymeric biomaterials capable of delivering nitric oxide (NO) topically can be used to enhance skin blood flow (SkBF) and accelerate wound healing. Herein, we used reversible addition-fragmentation chain transfer radical (RAFT) polymerization to synthesize the first poly(vinyl alcohol) (PVA) functionalized with terminal NO-releasing S-nitrosothiol (RSNO) groups for topical NO delivery. This strategy was based on the synthesis of a precursor amino-terminated PVA (PVA-NH), which was next functionalized with iminothiolane yielding 4-imino-4-amino-PVA-butane-1-thiol (PVA-SH), and finally S-nitrosated yielding S-nitroso 4-imino-4-amino-PVA-butane-1-thiol (PVA-SNO). Real-time chemiluminescence NO detection showed that blended films of pure PVA with PVA-SNO with mass ratios 30:70, 50:50 and 70:30 release NO with initial rates ranging from 1 to 12 nmol g min, and lead to a 2 to 10-fold dose-response increase in the SkBF, after topical application on the ventral forearm of volunteers. These results show that PVA-SNO is a potential platform for topical NO delivery in biomedical applications. 10.1016/j.niox.2020.03.002
Chitosan derivatives co-delivering nitric oxide and methicillin for the effective therapy to the methicillin-resistant S. aureus infection. Liu Shixin,Cai Xiang,Xue Wei,Ma Dong,Zhang Wu Carbohydrate polymers We developed a co-delivery system of nitric oxide (NO) and antibiotic for the antibiotic-resistant bacterial infection therapy. The NO could disperse the bacterial biofilms and convert the bacteria into an antibiotic-susceptible planktonic form. Using the chitosan-graft-poly(amidoamine) dendrimer (CS-PAMAM) as the co-delivery system, methicillin (MET) and NO were conjugated successively to form CS-PAMAM-MET/NONOate. The positive CS-PAMAM could efficiently capture the negatively charged bacteria and PAMAM provide abundant reaction points for high payloads of NO and MET. The CS-PAMAM-MET/NONOate displayed effective and combined antibacterial activity to the E. coli and S. aureus. Particularly, for the MET-resistant S. aureus (MRSA), the CS-PAMAM-MET/NONOate displayed the synergistic antibacterial activity. In vivo wound healing assays also confirmed that CS-PAMAM-MET/NONOate could heal the infection formed by MRSA and then accelerate the wound healing effectively. Moreover, CS-PAMAM-MET/NONOate showed no toxicity towards 3T3 cells in vitro and rats in vivo, providing a readily but high-efficient strategy to drug-resistant bacterial infection therapy. 10.1016/j.carbpol.2020.115928
S-Nitrosoglutathione loaded poly(lactic-co-glycolic acid) microparticles for prolonged nitric oxide release and enhanced healing of methicillin-resistant Staphylococcus aureus-infected wounds. Hlaing Shwe Phyu,Kim Jihyun,Lee Juho,Hasan Nurhasni,Cao Jiafu,Naeem Muhammad,Lee Eun Hee,Shin Jae Ho,Jung Yunjin,Lee Bok-Leul,Jhun Byung Hak,Yoo Jin-Wook European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V Methicillin-resistant Staphylococcus aureus (MRSA)-infected wounds have become a significant clinical issue worldwide. Recently, nitric oxide (NO) has emerged as a potent antibacterial agent against MRSA infections and a wound-healing enhancer. Nevertheless, clinical applications of NO have been largely restricted by its gaseous state and short half-life. In this study, our aim was to develop S-nitrosoglutathione (GSNO, an endogenous NO donor)-loaded poly(lactic-co-glycolic acid) [PLGA] microparticles (GSNO-MPs) that release NO over a prolonged period, to accelerate the healing of MRSA-infected wounds with less frequent dosing. GSNO was successfully encapsulated into PLGA microparticles by a solid-in-oil-in-water emulsion solvent evaporation method. Scanning electron microscopy and X-ray diffraction analyses confirmed the successful fabrication of GSNO-MPs. The latter released NO in a prolonged manner over 7 days and exerted a remarkable antibacterial activity against MRSA in a concentration- and time-dependent manner. Moreover, GSNO-MPs had good antibacterial efficacy and were found to accelerate wound healing in a mouse model of MRSA-infected wounds. Therefore, NO-releasing MPs devised in this study may be a promising option for the treatment of cutaneous wounds infected by drug-resistant bacteria such as MRSA. 10.1016/j.ejpb.2018.09.009
Nitric Oxide Chemical Donor Affects the Early Phases of In Vitro Wound Healing Process. La Torre Cristina,Cinque Benedetta,Lombardi Francesca,Miconi Gianfranca,Palumbo Paola,Evtoski Zoran,Placidi Giuseppe,Fanini Donatella,Cimini Anna Maria,Benedetti Elisabetta,Giuliani Maurizio,Cifone Maria Grazia Journal of cellular physiology An artificial wound in a confluent monolayer of human keratinocyte HaCaT cells or mouse embryo fibroblast Swiss NIH 3T3 cells was used to analyze the effects of the nitric oxide (NO) chemical donor, S-nitroso-N-acetylpenicillamine (SNAP). SNAP exposure promoted an enhanced rate of wound closure and accelerated motility of both keratinocytes and fibroblasts compared to control cells. The wounded monolayer cultures of HaCaT and NIH 3T3 cells, treated with or without SNAP, were monitored under a phase contrast microscope. Structural and ultrastructural modifications were analyzed by scanning electron microscopy (SEM). The images were captured by a digital camera at different time points (0-28 h) and the wound area was analyzed through software included in Matlab®. As early as 15 min, SNAP induced significant cytoskeletal remodeling, as shown by immunostaining (phalloidin-labelling), which in turn was associated with increased filopodium number and length rise. NO donor treatment also induced overexpression of Ki-67 protein, a typical marker of cell proliferation, as shown by immunostaining. Both SNAP-induced migration and proliferation were antagonized by the NO-sensitive GC inhibitor 1H-[1,2,4]oxadiazolo[-4,3-a]quinoxalin-1-one (ODQ), which suggests activation of the NO/cGMP signalling cascade in the observed SNAP-induced effects in the early stages of the healing process. Moreover, we provide evidence that PPAR-β antagonist (GSK0660) may interfere with NO-mediated wound healing process. J. Cell. Physiol. 231: 2185-2195, 2016. © 2016 Wiley Periodicals, Inc. 10.1002/jcp.25331
Chitosan-based nitric oxide-releasing dressing for anti-biofilm and in vivo healing activities in MRSA biofilm-infected wounds. Choi Moonjeong,Hasan Nurhasni,Cao Jiafu,Lee Juho,Hlaing Shwe Phyu,Yoo Jin-Wook International journal of biological macromolecules Bacterial biofilms on wounds impair the healing process and often lead to chronic wounds. Chitosan is a well-known biopolymer with antimicrobial and anti-biofilm effects. S-nitrosoglutathione (GSNO) has been identified as a promising nitric oxide (NO) donor to defend against pathogenic biofilms and enhance wound healing activities. In this study, we prepared NO-releasing chitosan film (CS/NO film) and evaluated its anti-biofilm activity and in vivo wound healing efficacy against methicillin-resistant Staphylococcus aureus (MRSA) biofilm-infected wounds in diabetic mice. The in vitro release study showed sustained release of NO over 3 days in simulated wound fluid. The CS/NO film significantly enhanced antibacterial activity against MRSA by > 3 logs reduction in bacterial viability. Moreover, CS/NO film exhibited a 3-fold higher anti-biofilm activity than the control and CS film. In in vivo MRSA biofilm-infected wounds, the CS/NO film-treated group showed faster biofilm dispersal, wound size reduction, epithelialization rates, and collagen deposition than the untreated and CS film-treated groups. Therefore, the CS/NO film investigated in this study could be a promising approach for the treatment of MRSA biofilm-infected wounds. 10.1016/j.ijbiomac.2019.10.009
Nitric oxide-releasing nanoparticles accelerate wound healing by promoting fibroblast migration and collagen deposition. Han George,Nguyen Long N,Macherla Chitralekha,Chi Yuling,Friedman Joel M,Nosanchuk Joshua D,Martinez Luis R The American journal of pathology Wound healing is a complex process that involves coordinated interactions between diverse immunological and biological systems. Long-term wounds remain a challenging clinical problem, affecting approximately 6 million patients per year, with a high economic impact. To exacerbate the problem, these wounds render the individual susceptible to life-threatening microbial infections. Because current therapeutic strategies have proved suboptimal, it is imperative to focus on new therapeutic approaches and the development of technologies for both short- and long-term wound management. In recent years, nitric oxide (NO) has emerged as a critical molecule in wound healing, with NO levels increasing rapidly after skin damage and gradually decreasing as the healing process progresses. In this study, we examined the effects of a novel NO-releasing nanoparticle technology on wound healing in mice. The results show that the NO nanoparticles (NO-np) significantly accelerated wound healing. NO-np modified leukocyte migration and increased tumor growth factor-β production in the wound area, which subsequently promoted angiogenesis to enhance the healing process. By using human dermal fibroblasts, we demonstrate that NO-np increased fibroblast migration and collagen deposition in wounded tissue. Together, these data show that NO-releasing nanoparticles have the ability to modulate and accelerate wound healing in a pleiotropic manner. 10.1016/j.ajpath.2011.12.013
The effects of topical nitric oxide on healing of partial thickness porcine burns. Singer Adam J,Choi Younghwan,Rashel Mohammed,Toussaint Jimmy,McClain Steve A Burns : journal of the International Society for Burn Injuries BACKGROUND:Nitric oxide is a wound mediator that promotes wound healing. We hypothesized that topical application of nitric oxide would speed reepithelialization, enhance angiogenesis, and reduce scar thickness in a partial thickness porcine burn model. METHODS:While under general anesthesia, 20 partial thickness burns were created on the backs of four female Yorkshire swine using a 2.5cm×2.5cm×7.5cm, 150-g aluminum bar, preheated to 80°C and applied for 20s. The necrotic epidermis was removed and the burns were randomized to low, medium, and high concentrations of a novel nitric-oxide (NO) releasing drug or its ointment vehicle applied 3 times weekly for 28 days. Full thickness punch biopsies were performed at 8, 11, 14 and 28 days after injury to determine percentage wound reepithelialization and scar thickness using H&E staining and blood vessel density using CD31 staining. RESULTS:At day 11, the percentages (SD) wound reepithelialization were: control, 26.3 (34.6); low NO, 23.9 (36.9); medium NO, 43.3 (42.9); and high NO, 59.9 (43.6); ANOVA, P=0.02. The number of CD31 stained blood vessels at days 8 and 11 were greater in wounds treated with high dose NO vs. controls (48.1 vs. 22.9 [P<0.001] and 44.0 vs. 33.5 [P=0.05] per 1mm respectively). Scar thicknesses (SD) in mm at day 28 by treatment allocation were: control, 4.8 (1.2); low NO, 4.7 (1.2); medium NO, 4.3 (1.2); and high NO, 4.1 (1.0); P=0.22. CONCLUSIONS:Treatment of partial thickness porcine burns with high concentrations of topical NO resulted in earlier reepithelization and increased angiogenesis but not reduced scar thickness compared with its control vehicle in a partial thickness porcine burn model. 10.1016/j.burns.2017.07.017
New generation of nitric oxide-releasing porous materials: Assessment of their potential to regulate biological functions. Pinto Rosana V,Fernandes Ana C,Antunes Fernando,Lin Zhi,Rocha João,Pires João,Pinto Moisés L Nitric oxide : biology and chemistry Nitric oxide (NO) presents innumerable biological roles, and its exogenous supplementation for therapeutic purposes has become a necessity. Some nanoporous materials proved to be potential vehicles for NO with high storage capacity. However, there is still a lack of information about their efficiency to release controlled NO and if they are biocompatible and biologically stable. In this work, we address this knowledge gap starting by evaluating the NO release and stability under biological conditions and their toxicity with primary keratinocyte cells. Titanosilicates (ETS-4 and ETS-10 types) and clay-based materials were the materials under study, which have shown in previous studies suitable NO gas adsorption/release rates. ETS-4 proved to be the most promising material, combining good biocompatibility at 180 μg/mL, stability and slower NO release. ETS-10 and ETAS-10 showed the best biocompatibility at the same concentration and, in the case of clay-based materials, CoOS is the least toxic of those tested and the one that releases the highest NO amount. The potentiality of these new NO donors to regulate biological functions was assessed next by controlling the mitochondrial respiration and the cell migration. NO-loaded ETS-4 regulates O consumption and cell migration in a dose-dependent manner. For cell migration, a biphasic effect was observed in a narrow range of ETS-4 concentration, with a stimulatory effect becoming inhibitory just by doubling ETS-4 concentration. For the other materials, no effective regulation was achieved, which highlights the relevance of the new assessment presented in this work for nanoporous NO carriers that will pave the way for further developments. 10.1016/j.niox.2019.05.010
Combined nitric oxide-releasing poly(vinyl alcohol) film/F127 hydrogel for accelerating wound healing. Schanuel Fernanda Seabra,Raggio Santos Karen Slis,Monte-Alto-Costa Andréa,de Oliveira Marcelo G Colloids and surfaces. B, Biointerfaces 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. 10.1016/j.colsurfb.2015.04.007
Nitric oxide-releasing chitosan film for enhanced antibacterial and in vivo wound-healing efficacy. Kim Jong Oh,Noh Jin-Ki,Thapa Raj Kumar,Hasan Nurhasni,Choi Moonjeong,Kim Jeong Hwan,Lee Joon-Hee,Ku Sae Kwang,Yoo Jin-Wook International journal of biological macromolecules Nitric oxide (NO) is a promising therapeutic agent with antibacterial and wound-healing properties. However, the gaseous state and short half-life of NO necessitate a formulation that can control its storage and release. In this study, we developed NO-releasing films (CS/NO film) composed of chitosan (CS) and S-nitrosoglutathione (GSNO) as a NO donor. Thermal analysis demonstrated molecular dispersion of GSNO in the films. In vitro release study revealed that NO release from CS/NO films followed Korsmeyer-Peppas model with Fickian diffusion kinetics. Moreover, the CS/NO film showed a stronger antibacterial activity against Pseudomonas aeruginosa (Gram-negative) and Staphylococcus aureus (Gram-positive) than the CS film. Further, the CS/NO film accelerated wound healing and epithelialization in a rat model of full-thickness wounds as compared to the CS film. Histopathological studies revealed that CS/NO films favorably enhanced the re-epithelialization and reconstruction of wounded skin. Therefore, our results suggest that CS/NO films could be a suitable formulation for treating full-thickness wounds. 10.1016/j.ijbiomac.2015.04.073
Nitric oxide-releasing poly(lactic-co-glycolic acid)-polyethylenimine nanoparticles for prolonged nitric oxide release, antibacterial efficacy, and in vivo wound healing activity. Nurhasni Hasan,Cao Jiafu,Choi Moonjeong,Kim Il,Lee Bok Luel,Jung Yunjin,Yoo Jin-Wook International journal of nanomedicine Nitric oxide (NO)-releasing nanoparticles (NPs) have emerged as a wound healing enhancer and a novel antibacterial agent that can circumvent antibiotic resistance. However, the NO release from NPs over extended periods of time is still inadequate for clinical application. In this study, we developed NO-releasing poly(lactic-co-glycolic acid)-polyethylenimine (PEI) NPs (NO/PPNPs) composed of poly(lactic-co-glycolic acid) and PEI/diazeniumdiolate (PEI/NONOate) for prolonged NO release, antibacterial efficacy, and wound healing activity. Successful preparation of PEI/NONOate was confirmed by proton nuclear magnetic resonance, Fourier transform infrared spectroscopy, and ultraviolet/visible spectrophotometry. NO/PPNPs were characterized by particle size, surface charge, and NO loading. The NO/PPNPs showed a prolonged NO release profile over 6 days without any burst release. The NO/PPNPs exhibited potent bactericidal efficacy against methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa concentration-dependently and showed the ability to bind on the surface of the bacteria. We also found that the NO released from the NO/PPNPs mediates bactericidal efficacy and is not toxic to healthy fibroblast cells. Furthermore, NO/PPNPs accelerated wound healing and epithelialization in a mouse model of a MRSA-infected wound. Therefore, our results suggest that the NO/PPNPs presented in this study could be a suitable approach for treating wounds and various skin infections. 10.2147/IJN.S82199
Activity of a nitric oxide-generating wound treatment system against wound pathogen biofilms. Waite Richard D,Stewart Joanne E,Stephen Abish S,Allaker Robert P International journal of antimicrobial agents Wound bioburden plays an important role in impaired healing and development of infection-related complications. The objective of this study was to determine the efficacy of an innovative two-layer nitric oxide-generating system (NOx) to prevent and treat biofilms formed by bacterial and fungal pathogens commonly associated with wound infection, and activity against Pseudomonas aeruginosa virulence factors. Single- and mixed-species biofilms were grown for 24 h on nitrocellulose filters placed on agar. Filters were covered with either NOx or placebo, before and after biofilm formation. Populations of bacteria and yeasts were determined using viable counts. Pyocyanin and elastase production from P. aeruginosa were determined in supernatants derived from suspended biofilms. Efficacy of NOx was demonstrated against Staphylococcus aureus, P. aeruginosa, Acinetobacter baumannii, Escherichia coli and Candida spp. Population reductions between 2- and 10-log fold were observed. Pyocyanin and elastase activities from P. aeruginosa were reduced 1.9- and 3.2-fold, respectively. This study demonstrated activity of NOx against formation and treatment of single- and mixed-species biofilms, including multidrug-resistant strains. NOx represents a new generation of antimicrobial agent with potent, broad-spectrum activity, and with no evidence of resistance development. 10.1016/j.ijantimicag.2018.04.009
Nitric oxide releasing two-part creams containing S-nitrosoglutathione and zinc oxide for potential topical antimicrobial applications. Doverspike Joshua C,Zhou Yang,Wu Jianfeng,Tan Xiaojuan,Xi Chuanwu,Meyerhoff Mark E Nitric oxide : biology and chemistry Currently, most antimicrobial topical treatments utilize antibiotics to prevent or treat infection at a wound site. However, with the ongoing evolution of multi-drug resistant bacterial strains, there is a high demand for alternative antimicrobial treatments. Nitric oxide (NO) is an endogenous gas molecule with potent antimicrobial activity, which is effective against a wide variety of bacterial strains. In this study, the potential for creating NO releasing creams containing the naturally occurring NO carrier, S-nitrosoglutathione (GSNO), are characterized and evaluated. GSNO is shown to have prolonged stability (>300 days) when mixed and stored within Vaseline at 24 °C. Further, enhanced proliferation of NO from GSNO using zinc oxide nanoparticles (ZnO) is demonstrated. Triggering NO release from the GSNO/Vaseline mixture using a commercial zinc oxide-containing cream exhibits first-order NO release kinetics with the highest %NO release over the first 6 h. Significant killing effects against S. aureus, S. epidermidis, and P. aeruginosa are demonstrated for the GSNO/Vaseline/ZnO cream mixtures in a proportional manner dependent upon the concentration of GSNO in the final mixture. 10.1016/j.niox.2019.05.009
Hollow, Rough, and Nitric Oxide-Releasing Cerium Oxide Nanoparticles for Promoting Multiple Stages of Wound Healing. Ma Xiaomin,Cheng Yan,Jian Hui,Feng Yanlin,Chang Yun,Zheng Runxiao,Wu Xiaqing,Wang Li,Li Xi,Zhang Haiyuan Advanced healthcare materials Wound healing is a complex and sequential biological process that involves multiple stages. Although various nanomaterials are applied to accelerate the wound healing process, only a single stage is promoted during the process, lacking hierarchical stimulation. Herein, hollow CeO nanoparticles (NPs) with rough surface and l-arginine inside ( CeO NPs) are developed as a compact and programmable nanosystem for sequentially promoting the hemostasis, inflammation, and proliferation stages. The rough surface of CeO NPs works as a nanobridge to rapidly closure the wounds, promoting the hemostasis stage. The hollow structure of CeO NPs enables the multireflection of light inside particles, significantly enhancing the light harvest efficiency and electron-hole pair abundance. Simultaneously, the porous shell of CeO NPs facilitates the electron-hole separation and reactive oxygen species production, preventing wound infection and promotion wound healing during the inflammation stage. The enzyme mimicking property of CeO NPs can alleviate the oxidative injury in the wound, and the released l-arginine can be converted into nitric oxide (NO) under the catalysis of inducible NO synthase, both of which promote the proliferation stage. A series of in vitro and in vitro biological assessments corroborate the effectiveness of CeO NPs in the wound healing process. 10.1002/adhm.201900256
Nitric oxide-releasing polymer incorporated ointment for cutaneous wound healing. Kang Youngnam,Kim Jihoon,Lee Yeong Mi,Im Sooseok,Park Hansoo,Kim Won Jong Journal of controlled release : official journal of the Controlled Release Society This work demonstrates the development of nitric oxide-releasing ointment and its potential on efficient wound healing. Nitric oxide-releasing polymer was successfully synthesized, which is composed of biocompatible Pluronic F127, branched polyethylenimine and 1-substituted diazen-1-ium-1,2-diolates. The synthesized nitric oxide-releasing polymer was incorporated into the PEG-based ointment which not only facilitated nitric oxide release in a slow manner, but also served as a moisturizer to enhance the wound healing. As compared to control groups, the nitric oxide-releasing ointment showed the accelerated wound closure with enhanced re-epithelialization, collagen deposition, and blood vessel formation in vivo. Therefore, this nitric oxide-based ointment presents the promising potential for the efficient strategy to heal the cutaneous wound. 10.1016/j.jconrel.2015.08.057
Antimicrobial efficacy and wound-healing property of a topical ointment containing nitric-oxide-loaded zeolites. Neidrauer Michael,Ercan Utku K,Bhattacharyya Aparna,Samuels Joshua,Sedlak Jason,Trikha Ritika,Barbee Kenneth A,Weingarten Michael S,Joshi Suresh G Journal of medical microbiology Topical delivery of nitric oxide (NO) through a wound dressing has the potential to reduce wound infections and improve healing of acute and chronic wounds. This study characterized the antibacterial efficacy of an ointment containing NO-loaded, zinc-exchanged zeolite A that releases NO upon contact with water. The release rate of NO from the ointment was measured using a chemiluminescence detection system. Minimum bactericidal concentration assays were performed using five common wound pathogens, including Gram-negative bacteria (Escherichia coli and Acinetobacter baumannii), Gram-positive bacteria (Staphylococcus epidermidis and meticillin-resistant Staphylococcus aureus) and a fungus (Candida albicans). The time dependence of antimicrobial activity was characterized by performing log-reduction assays at four time points after 1-8 h ointment exposure. The cytotoxicity of the ointment after 24 h was assessed using cultured 3T3 fibroblast cells. Minimum microbicidal concentrations (MMCs) for bacterial organisms (5×10(7) c.f.u.) ranged from 50 to 100 mg ointment (ml media)(-1); the MMC for C. albicans (5×10(4) c.f.u.) was 50 mg ointment (ml media)(-1). Five to eight log reductions in bacterial viability and three log reductions in fungal viability were observed after 8 h exposure to NO-zeolite ointment compared with untreated organisms. Fibroblasts remained viable after 24 h exposure to the same concentration of NO-zeolite ointment as was used in antimicrobial tests. In parallel studies, full-thickness cutaneous wounds on Zucker obese rats healed faster than wounds treated with a control ointment. These data indicate that ointment containing NO-loaded zeolites could potentially be used as a broad-spectrum antimicrobial wound-healing dressing. 10.1099/jmm.0.067322-0
Copper-Based Metal-Organic Framework as a Controllable Nitric Oxide-Releasing Vehicle for Enhanced Diabetic Wound Healing. Zhang Pengju,Li You,Tang Yaohui,Shen Hui,Li Jiankai,Yi Zhengfang,Ke Qinfei,Xu He ACS applied materials & interfaces Chronic wounds are one of the most serious complications of diabetes mellitus. Even though utilizing nitric oxide (NO) as a gas medicine to repair diabetic wounds presents a promising strategy, controlling the NO release behavior in the affected area, which is vital for NO-based therapy, still remains a significant challenge. In this work, a copper-based metal-organic framework, namely, HKUST-1, has been introduced as a NO-loading vehicle, and a NO sustained release system with the core-shell structure has been designed through the electrospinning method. The results show that the NO is quantificationally and stably loaded in the HKUST-1 particles, and the NO-loaded HKUST-1 particles are well incorporated into the core layer of the coaxial nanofiber. Therefore, NO can be controllably released with an average release rate of 1.74 nmol L h for more than 14 days. Moreover, the additional copper ions released from the degradable HKUST-1 play a synergistic role with NO to promote endothelial cell growth and significantly improve the angiogenesis, collagen deposition as well as anti-inflammatory property in the wound bed, which eventually accelerate the diabetic wound healing. These results suggest that such a copper-based metal-organic framework material as a controllable NO-releasing vehicle is a highly efficient therapy for diabetic wounds. 10.1021/acsami.0c01792