Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Vasc Interv Radiol ; 32(6): 882-889, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33689833

RESUMEN

PURPOSE: To compare the performance of a dual-lumen flushable drainage catheter to a conventional catheter for complex fluid collection drainage. METHODS: Two prototype catheters (20- and 28-F) were created by incorporating a customized infusion lumen within the wall of a large-bore conventional drainage catheter, which facilitated simultaneous irrigation of the drainage lumen and the targeted collection via inward- and outward-facing infusion side holes. These were tested against unaltered 20- and 28-F conventional catheters to determine if the injection of a dedicated flush lumen improved rapidity and completeness of gravity drainage. In vitro models were created to simulate serous fluid, purulent/exudative fluid, particulate debris, and acute hematoma. RESULTS: In the purulent model, mean drainage rate was 19.9 ± 8.0 and 9.5±1.4 mL/min for the 20-F prototype and control (P < .001) and 63.9 ± 4.3 and 35.4 ± 3.4 mL/min for the 28-F prototype and control (P = .006), respectively, with complete drainage achieved in all trials. In the particulate model, mean drainage rate was 24.5 ± 9.7 and 12.0 ± 12.5 mL/min for the 28-F prototype and control (P = .003), respectively, with 69.0% versus 41.1% total drainage achieved over 24 minutes (P = .029). In the hematoma model, mean drainage rate was 22.7 ± 4.6 and 4.8 ± 4.3 mL/min for the 28-F prototype and control (P = .022), respectively, with 80.3% versus 20.1% drainage achieved over 15 minutes (P = .003). Particulate and hematoma 20-F prototypes and conventional trials failed due to immediate occlusion. CONCLUSIONS: The proposed dual-lumen drainage catheter with irrigation of a dedicated flush lumen improved evacuation of complex fluid collections in vitro.


Asunto(s)
Cateterismo/instrumentación , Catéteres , Drenaje/instrumentación , Irrigación Terapéutica/instrumentación , Diseño de Equipo , Ensayo de Materiales , Factores de Tiempo
2.
Small ; 16(29): e2000171, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32529791

RESUMEN

Cell separation is a key step in many biomedical research areas including biotechnology, cancer research, regenerative medicine, and drug discovery. While conventional cell sorting approaches have led to high-efficiency sorting by exploiting the cell's specific properties, microfluidics has shown great promise in cell separation by exploiting different physical principles and using different properties of the cells. In particular, label-free cell separation techniques are highly recommended to minimize cell damage and avoid costly and labor-intensive steps of labeling molecular signatures of cells. In general, microfluidic-based cell sorting approaches can separate cells using "intrinsic" (e.g., fluid dynamic forces) versus "extrinsic" external forces (e.g., magnetic, electric field, etc.) and by using different properties of cells including size, density, deformability, shape, as well as electrical, magnetic, and compressibility/acoustic properties to select target cells from a heterogeneous cell population. In this work, principles and applications of the most commonly used label-free microfluidic-based cell separation methods are described. In particular, applications of microfluidic methods for the separation of circulating tumor cells, blood cells, immune cells, stem cells, and other biological cells are summarized. Computational approaches complementing such microfluidic methods are also explained. Finally, challenges and perspectives to further develop microfluidic-based cell separation methods are discussed.


Asunto(s)
Técnicas Analíticas Microfluídicas , Células Neoplásicas Circulantes , Recuento de Células , Separación Celular , Humanos , Microfluídica
3.
Small ; 16(25): e2001837, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32419312

RESUMEN

Stem cells secrete trophic factors that induce angiogenesis. These soluble factors are promising candidates for stem cell-based therapies, especially for cardiovascular diseases. Mechanical stimuli and biophysical factors presented in the stem cell microenvironment play important roles in guiding their behaviors. However, the complex interplay and precise role of these cues in directing pro-angiogenic signaling remain unclear. Here, a platform is designed using gelatin methacryloyl hydrogels with tunable rigidity and a dynamic mechanical compression bioreactor to evaluate the influence of matrix rigidity and mechanical stimuli on the secretion of pro-angiogenic factors from human mesenchymal stem cells (hMSCs). Cells cultured in matrices mimicking mechanical elasticity of bone tissues in vivo show elevated secretion of vascular endothelial growth factor (VEGF), one of representative signaling proteins promoting angiogenesis, as well as increased vascularization of human umbilical vein endothelial cells (HUVECs) with a supplement of conditioned media from hMSCs cultured across different conditions. When hMSCs are cultured in matrices stimulated with a range of cyclic compressions, increased VEGF secretion is observed with increasing mechanical strains, which is also in line with the enhanced tubulogenesis of HUVECs. Moreover, it is demonstrated that matrix stiffness and cyclic compression modulate secretion of pro-angiogenic molecules from hMSCs through yes-associated protein activity.


Asunto(s)
Células Madre Mesenquimatosas , Células Cultivadas , Señales (Psicología) , Medios de Cultivo Condicionados , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular
4.
Biotechnol Bioeng ; 117(7): 2237-2246, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32215917

RESUMEN

The chances of ventilator-associated pneumonia (VAP) increases 6-20 folds when an endotracheal tube (ETT) is placed in a patient. VAP is one of the most common hospital-acquired infections and comprises 86% of the nosocomial pneumonia cases. This study introduces the idea of nitric oxide-releasing ETTs (NORel-ETTs) fabricated by the incorporation of the nitric oxide (NO) donor S-nitroso-N-acetylpenicillamine (SNAP) into commercially available ETTs via solvent swelling. The impregnation of SNAP provides NO release over a 7-day period without altering the mechanical properties of the ETT. The NORel-ETTs successfully reduced the bacterial infection from a commonly found pathogen in VAP, Pseudomonas aeruginosa, by 92.72 ± 0.97% when compared with the control ETTs. Overall, this study presents the incorporation of the active release of a bactericidal agent in ETTs as an efficient strategy to prevent the risk of VAP.


Asunto(s)
Antibacterianos/administración & dosificación , Intubación Intratraqueal/instrumentación , Donantes de Óxido Nítrico/administración & dosificación , Neumonía Asociada al Ventilador/prevención & control , S-Nitroso-N-Acetilpenicilamina/administración & dosificación , Antibacterianos/farmacología , Liberación de Fármacos , Humanos , Intubación Intratraqueal/métodos , Donantes de Óxido Nítrico/farmacología , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación , S-Nitroso-N-Acetilpenicilamina/farmacología
5.
J Biomed Mater Res A ; 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769626

RESUMEN

Wound infection and excessive blood loss are the two major challenges associated with trauma injuries that account for 10% of annual deaths in the United States. Nitric oxide (NO) is a gasotransmitter cell signaling molecule that plays a crucial role in the natural wound healing process due to its antibacterial, anti-inflammatory, cell proliferation, and tissue remodeling abilities. Tranexamic acid (TXA), a prothrombotic agent, has been used topically and systemically to control blood loss in reported cases of epistaxis and combat-related trauma injuries. Its properties could be incorporated in wound dressings to induce immediate clot formation, which is a key factor in controlling excessive blood loss. This study introduces a novel, instant clot-forming NO-releasing dressing, and fabricated using a strategic bi-layer configuration. The layer adjacent to the wound was designed with TXA suspended on a resinous bed of propolis, which is a natural bioadhesive possessing antibacterial and anti-inflammatory properties. The base layer, located furthest away from the wound, has an NO donor, S-nitroso-N-acetylpenicillamine (SNAP), embedded in a polymeric bed of Carbosil®, a copolymer of polycarbonate urethane and silicone. Propolis was integrated with a uniform layer of TXA in variable concentrations: 2.5, 5.0, and 7.5 vol % of propolis. This design of the TXA-SNAP-propolis (T-SP) wound dressing allows TXA to form a more stable clot by preventing the lysis of fibrin. The lactate dehydrogenase-based platelet adhesion assay showed an increase in fibrin activation with 7.5% T-SP as compared with control within the first 15 min of its application. A scanning electron microscope (SEM) confirmed the presence of a dense fibrin network stabilizing the clot for fabricated dressing. The antibacterial activity of NO and propolis resulted in a 98.9 ± 1% and 99.4 ± 1% reduction in the colony-forming unit of Staphylococcus aureus and multidrug-resistant Acinetobacter baumannii, respectively, which puts forward the fabricated dressing as an emergency first aid for traumatic injuries, preventing excessive blood loss and soil-borne infections.

6.
ACS Appl Mater Interfaces ; 14(9): 11116-11123, 2022 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-35225600

RESUMEN

Blood-contacting medical devices (BCMDs) are inevitably challenged by thrombi formation, leading to occlusion of flow and device failure. Ideal BCMDs seek to mimic the intrinsic antithrombotic properties of the human vasculature to locally prevent thrombotic complications, negating the need for systemic anticoagulation. An emerging category of BCMD technology utilizes nitric oxide (NO) as a hemocompatible agent, as the vasculature's endothelial layer naturally releases NO to inhibit platelet activation and consumption. In this paper, we report for the first time the novel impregnation of S-nitrosoglutathione (GSNO) into polymeric poly(vinyl chloride) (PVC) tubing via an optimized solvent-swelling method. Material testing revealed an optimized GSNO-PVC material that had adequate GSNO loading to achieve NO flux values within the physiological endothelial NO flux range for a 4 h period. Through in vitro hemocompatibility testing, the optimized material was deemed nonhemolytic (hemolytic index <2%) and capable of reducing platelet activation, suggesting that the material is suitable for contact with whole blood. Furthermore, an in vivo 4 h extracorporeal circulation (ECC) rabbit thrombogenicity model confirmed the blood biocompatibility of the optimized GSNO-PVC. Platelet count remained near 100% for the novel GSNO-impregnated PVC loops (1 h, 91.08 ± 6.27%; 2 h, 95.68 ± 0.61%; 3 h, 97.56 ± 8.59%; 4 h, 95.11 ± 8.30%). In contrast, unmodified PVC ECC loops occluded shortly after the 2 h time point and viable platelet counts quickly diminished (1 h, 85.67 ± 12.62%; 2 h, 54.46 ± 10.53%; 3 h, n/a; 4 h, n/a). The blood clots for GSNO-PVC loops (190.73 ± 72.46 mg) compared to those of unmodified PVC loops (866.50 ± 197.98 mg) were significantly smaller (p < 0.01). The results presented in this paper recommend further investigation in long-term animal models and suggest that GSNO-PVC has the potential to serve as an alternative to systemic anticoagulation in BCMD applications.


Asunto(s)
Polímeros/farmacología , S-Nitrosoglutatión/farmacología , Animales , Coagulación Sanguínea/efectos de los fármacos , Circulación Extracorporea/métodos , Hemólisis/efectos de los fármacos , Masculino , Ensayo de Materiales , Modelos Animales , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacología , Activación Plaquetaria/efectos de los fármacos , Polímeros/uso terapéutico , Cloruro de Polivinilo/química , Conejos , S-Nitrosoglutatión/química , S-Nitrosoglutatión/uso terapéutico , Propiedades de Superficie , Porcinos , Trombosis/prevención & control
7.
ACS Biomater Sci Eng ; 7(11): 5279-5287, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34695358

RESUMEN

Attachment of a nitric oxide (NO) donor to an electrospun polymer has the potential to improve its proliferative and antimicrobial capabilities. This study presents the novel, covalent attachment of S-nitroso-N-acetylpenicillamine (SNAP) to polyacrylonitrile (PAN) fibers. By attaching the NO donor to the polymer, rather than blending it, leaching is reduced to maintain a NO flux within the physiologically relevant range for a longer duration, while limiting any cytotoxic effects. The synthesized fibers were characterized using a variety of techniques such as scanning electron microscopy, 1H NMR, and drop shape analysis. Due to the antimicrobial activity of NO, the SNAP-PAN fibers demonstrated a 2-log reduction of S. aureus adhesion. Furthermore, the extended zone of inhibition of S. aureus by SNAP-PAN demonstrates the ability of NO to impact the environment surrounding the material, in addition to the environment in direct contact with it. The combination of NO release, hydrophilicity of PAN, and the fibrous network led to increased fibroblast proliferation and attachment, potentially expanding the fibers as an improved cell scaffolding platform. The results from this study demonstrate a novel preparation and design of NO-releasing fibers to provide multiple benefits for a variety of biomedical applications.


Asunto(s)
Nanofibras , Resinas Acrílicas , S-Nitroso-N-Acetilpenicilamina/farmacología , Staphylococcus aureus , Ingeniería de Tejidos
8.
ACS Appl Bio Mater ; 3(11): 7677-7686, 2020 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-35019507

RESUMEN

Blended and coaxial fibers comprising polycaprolactone and gelatin, containing the endogenous nitric oxide (NO) donor S-nitrosoglutathione (GSNO), were electrospun. Both types of fibers had their NO release profiles tested under physiological conditions to examine their potential applications as biomedical scaffolds. The coaxial fibers exhibited a prolonged and consistent release of NO over the course of 4 d from the core-encapsulated GSNO, while the blended fibers had a large initial release and leaching of GSNO that was exhausted over a shorter period of time. Bacterial testing of both fiber scaffolds was conducted over a 24 h period against Staphylococcus aureus (S. aureus) and demonstrated a 3-log reduction in bacterial viability. In addition, no cytotoxic response was reported when the material was tested on mouse fibroblast cells in vitro. These fibrous matrices were also shown to support cell growth, attachment, and overall activity of fibroblasts when exposed to NO, especially when GSNO was encapsulated within coaxial fibers. From an application point of view, these NO-releasing fibers offer great potential in tissue engineering and biomedical applications because of the crucial role of NO in regulating a variety of biological processes in humans such as angiogenesis, tissue remodeling, and eliminating foreign pathogens.

9.
ACS Appl Mater Interfaces ; 12(18): 20158-20171, 2020 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-32271542

RESUMEN

The management of thrombosis and bacterial infection is critical to ensure the functionality of medical devices. While administration of anticoagulants is the current antithrombotic clinical practice, a variety of complications, such as uncontrolled hemorrhages or heparin-induced thrombocytopenia, can occur. Additionally, infection rates remain a costly and deadly complication associated with use of these medical devices. It has been hypothesized that if a synthetic surface could mimic the biochemical mechanisms of the endothelium of blood vessels, thrombosis could be reduced, anticoagulant use could be avoided, and infection could be prevented. Herein, the interfacial biochemical effects of the endothelium were mimicked by altering the surface of medical grade silicone rubber (SR). Surface modification was accomplished via heparin surface immobilization (Hep) and the inclusion of a nitric oxide (NO) donor into the SR polymeric matrix to achieve synergistic effects (Hep-NO-SR). An in vitro bacteria adhesion study revealed that Hep-NO-SR exhibited a 99.46 ± 0.17% reduction in viable bacteria adhesion compared to SR. An in vitro platelet study revealed Hep-NO-SR reduced platelet adhesion by 84.12 ± 6.19% compared to SR, while not generating a cytotoxic response against fibroblast cells. In a 4 h extracorporeal circuit model without systemic anticoagulation, all Hep-NO-SR samples were able to maintain baseline platelet count and device patency; whereas 66% of SR samples clotted within the first 2 h of study. Results indicate that Hep-NO-SR creates a more hemocompatible and antibacterial surface by mimicking two key biochemical functions of the native endothelium.


Asunto(s)
Materiales Biomiméticos/química , Fármacos Hematológicos/uso terapéutico , Heparina/uso terapéutico , Donantes de Óxido Nítrico/uso terapéutico , S-Nitroso-N-Acetilpenicilamina/uso terapéutico , Animales , Adhesión Bacteriana/efectos de los fármacos , Materiales Biomiméticos/toxicidad , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/toxicidad , Endotelio/química , Fármacos Hematológicos/farmacología , Fármacos Hematológicos/toxicidad , Heparina/farmacología , Heparina/toxicidad , Proteínas Inmovilizadas/farmacología , Proteínas Inmovilizadas/uso terapéutico , Proteínas Inmovilizadas/toxicidad , Ratones , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/toxicidad , Adhesividad Plaquetaria/efectos de los fármacos , Conejos , S-Nitroso-N-Acetilpenicilamina/farmacología , S-Nitroso-N-Acetilpenicilamina/toxicidad , Elastómeros de Silicona/química , Elastómeros de Silicona/toxicidad , Staphylococcus aureus/efectos de los fármacos , Propiedades de Superficie
10.
Artículo en Inglés | MEDLINE | ID: mdl-32528214

RESUMEN

The detection of cardiac troponin I (cTnI) is clinically used to monitor myocardial infarctions (MI) and other heart diseases. The development of highly sensitive detection assays for cTnI is needed for the efficient diagnosis and monitoring of cTnI levels. Traditionally, enzyme-based immunoassays have been used for the detection of cTnI. However, the use of label-free sensing techniques have the advantage of potentially higher speed and lower cost for the assays. We previously reported a Photonic Crystal-Total Internal Reflection (PC-TIR) biosensor for label-free quantification of cTnI. To further improve on this, we present a comparative study between an antibody based PC-TIR sensor that relies on recombinant protein G (RPG) for the proper orientation of anti-cTnI antibodies, and an aptamer-based PC-TIR sensor for improved sensitivity and performance. Both assays relied on the use of polyethylene glycol (PEG) linkers to facilitate the modification of the sensor surfaces with biorecognition elements and to provide fluidity of the sensing surface. The aptamer-based PC-TIR sensor was successfully able to detect 0.1 ng/mL of cTnI. For the antibody-based PC-TIR sensor, the combination of the fluidity of the PEG and the increased number of active antibodies allowed for an improvement in assay sensitivity with a low detection limit of 0.01 ng/mL. The developed assays showed good performance and potential to be applied for the detection of cTnI levels in clinical samples upon further development.

11.
Biomaterials ; 255: 120196, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32623181

RESUMEN

Organ-on-a-chip technology tries to mimic the complexity of native tissues in vitro. Important progress has recently been made in using this technology to study the gut with and without microbiota. These in vitro models can serve as an alternative to animal models for studying physiology, pathology, and pharmacology. While these models have greater physiological relevance than two-dimensional (2D) cell systems in vitro, endocrine and immunological functions in gut-on-a-chip models are still poorly represented. Furthermore, the construction of complex models, in which different cell types and structures interact, remains a challenge. Generally, gut-on-a-chip models have the potential to advance our understanding of the basic interactions found within the gut and lay the foundation for future applications in understanding pathophysiology, developing drugs, and personalizing medical treatments.


Asunto(s)
Microbioma Gastrointestinal , Dispositivos Laboratorio en un Chip , Animales
12.
ACS Appl Mater Interfaces ; 12(8): 9070-9079, 2020 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-32009376

RESUMEN

The development of nonfouling and antimicrobial materials has shown great promise for reducing thrombosis and infection associated with medical devices with aims of improving device safety and decreasing the frequency of antibiotic administration. Here, the design of an antimicrobial, anti-inflammatory, and antithrombotic vascular catheter is assessed in vivo over 7 d in a rabbit model. Antimicrobial and antithrombotic activity is achieved through the integration of a nitric oxide donor, while the nonfouling surface is achieved using a covalently bound phosphorylcholine-based polyzwitterionic copolymer topcoat. The effect of sterilization on the nonfouling nature and nitric oxide release is presented. The catheters reduced viability of Staphylococcus aureus in long-term studies (7 d in a CDC bioreactor) and inflammation in the 7 d rabbit model. Overall, this approach provides a robust method for decreasing thrombosis, inflammation, and infections associated with vascular catheters.


Asunto(s)
Antibacterianos , Infecciones Relacionadas con Catéteres/prevención & control , Catéteres , Materiales Biocompatibles Revestidos , Óxido Nítrico , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/crecimiento & desarrollo , Trombosis/prevención & control , Animales , Antibacterianos/química , Antibacterianos/farmacología , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Óxido Nítrico/química , Óxido Nítrico/farmacología , Conejos
13.
Small Methods ; 4(1)2020 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-33043130

RESUMEN

Microphysiological systems, also known as organ-on-a-chip platforms, show promise for the development of new testing methods that can be more accurate than both conventional two-dimensional cultures and costly animal studies. The development of more intricate microphysiological systems can help to better mimic the human physiology and highlight the systemic effects of different drugs and materials. Nanomaterials are among a technologically important class of materials used for diagnostic, therapeutic, and monitoring purposes; all of which and can be tested using new organ-on-a-chip systems. In addition, the toxicity of nanomaterials which have entered the body from ambient air or diet can have deleterious effects on various body systems. This in turn can be studied in newly developed microphysiological systems. While organ-on-a-chip models can be useful, they cannot pick up secondary and systemic toxicity. Thus, the utilization of multi-organ-on-a-chip systems for advancing nanotechnology will largely be reflected in the future of drug development, toxicology studies and precision medicine. Various aspects of related studies, current challenges, and future perspectives are discussed in this paper.

14.
Acta Biomater ; 112: 190-201, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32434076

RESUMEN

Microfluidic artificial lungs (µALs) have the potential to improve the treatment and quality of life for patients with acute or chronic lung injury. In order to realize the full potential of this technology (including as a destination therapy), the biocompatibility of these devices needs to be improved to produce long-lasting devices that are safe for patient use with minimal or no systemic anticoagulation. Many studies exist which probe coagulation and thrombosis on polydimethyl siloxane (PDMS) surfaces, and many strategies have been explored to improve surface biocompatibility. As the field of µALs is young, there are few studies which investigate biocompatibility of functioning µALs; and even fewer which were performed in vivo. Here, we use both in vitro and in vivo models to investigate two strategies to improve µAL biocompatibility: 1) a hydrophilic surface coating (polyethylene glycol, PEG) to prevent surface fouling, and 2) the addition of nitric oxide (NO) to the sweep gas to inhibit platelet activation locally within the µAL. In this study, we challenge µALs with clottable blood or platelet-rich plasma (PRP) and monitor the resistance to blood flow over time. Device lifetime (the amount of time the µAL remains patent and unobstructed by clot) is used as the primary indicator of biocompatibility. This study is the first study to: 1) investigate the effect of NO release on biocompatibility in a microfluidic network; 2) combine a hydrophilic PEG coating with NO release to improve blood compatibility; and 3) perform extended in vivo biocompatibility testing of a µAL. We found that µALs challenged in vitro with PRP remained patent significantly longer when the sweep gas contained NO than without NO. In the in vivo rabbit model, neither approach alone (PEG coating nor NO sweep gas) significantly improved biocompatibility compared to controls (though with larger sample size significance may become apparent); while the combination of a PEG coating with NO sweep gas resulted in significant improvement of device lifetime. STATEMENT OF SIGNIFICANCE: The development of microfluidic artificial lungs (µALs) can potentially have a massive impact on the treatment of patients with acute and chronic lung impairments. Before these devices can be deployed clinically, the biocompatibility of µALs must be improved and more comprehensively understood. This work explores two strategies for improving biocompatibility, a hydrophilic surface coating (polyethylene glycol) for general surface passivation and the addition of nitric oxide (NO) to the sweep gas to quell platelet and leukocyte activation. These two strategies are investigated separately and as a combined device treatment. Devices are challenged with clottable blood using in vitro testing and in vivo testing in rabbits. This is the first study to our knowledge that allows statistical comparisons of biocompatible µALs in animals, a key step towards eventual clinical use.


Asunto(s)
Microfluídica , Calidad de Vida , Animales , Plaquetas , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Pulmón , Conejos
15.
Adv Drug Deliv Rev ; 165-166: 41-59, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31837356

RESUMEN

Microneedles (MNs) have been used to deliver drugs for over two decades. These platforms have been proven to increase transdermal drug delivery efficiency dramatically by penetrating restrictive tissue barriers in a minimally invasive manner. While much of the early development of MNs focused on transdermal drug delivery, this technology can be applied to a variety of other non-transdermal biomedical applications. Several variations, such as multi-layer or hollow MNs, have been developed to cater to the needs of specific applications. The heterogeneity in the design of MNs has demanded similar variety in their fabrication methods; the most common methods include micromolding and drawing lithography. Numerous materials have been explored for MN fabrication which range from biocompatible ceramics and metals to natural and synthetic biodegradable polymers. Recent advances in MN engineering have diversified MNs to include unique shapes, materials, and mechanical properties that can be tailored for organ-specific applications. In this review, we discuss the design and creation of modern MNs that aim to surpass the biological barriers of non-transdermal drug delivery in ocular, vascular, oral, and mucosal tissue.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Microinyecciones/instrumentación , Microinyecciones/métodos , Administración Tópica , Transporte Biológico , Diseño de Equipo , Humanos , Microtecnología/métodos , Polímeros , Prótesis e Implantes
16.
ACS Biomater Sci Eng ; 5(4): 2021-2029, 2019 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33405516

RESUMEN

Urinary catheterization is one of the most common medical procedures that makes a patient susceptible to infection due to biofilm formation on the urinary catheter. Catheter associated urinary tract infections (CAUTIs) are responsible for over 1 million cases in the United States alone and cost the healthcare industry more than $350 million every year. This work presents a liquid-infused nitric-oxide-releasing (LINORel) urinary catheter fabricated by incorporating the nitric oxide (NO) donor S-nitroso-N-acetylpenicillamine (SNAP) and silicone oil into commercial silicone Foley catheters through a two-stage swelling process. This synergistic combination improves NO-releasing materials by providing minimal SNAP leaching and a more controlled release of NO while incorporating the nonfouling characteristics of liquid-infused materials. The LINORel urinary catheter was successful in sustaining a controlled NO release over a 60 day period under physiological conditions with minimal SNAP leaching during the initial 24 h period, 0.49 ± 0.0061%. The LINORel-UC proved successful in reducing bacterial adhesion and biofilm formation for Gram positive Staphylococcus aureus (98.49 ± 2.06%) over a 7 day period in a drip flow bioreactor environment. Overall, this study presents a desirable combination that incorporates the antifouling advantages of liquid-infused materials with the active release of a bactericidal agent, an uncharted territory in aiding to prevent the risk of CAUTIs.

17.
J Biomed Mater Res B Appl Biomater ; 107(4): 1068-1078, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30230685

RESUMEN

Open bone fractures are not only difficult to heal but also are at a high risk of infections. Annual cases of fractures which result from osteoporosis amount to approximately 9 million. Endogenously released nitric oxide (NO) has been shown to play a role in osteogenic differentiation in addition to eradicating infection against a wide variety of pathogens. In the current work, antimicrobial NO releasing 3D bone scaffolds were fabricated using S-nitroso-N-acetyl-penicillamine (SNAP) as the NO donor. During fabrication, nano-hydroxyapatite (nHA) was added to each of the scaffolds in the concentration range of 10-50 wt % in nHA-starch-alginate and nHA-starch-chitosan scaffolds. The mechanical strength of the scaffolds increased proportionally to the concentration of nHA and 50 wt % nHA-starch-alginate possessed the highest load bearing capacity of 203.95 ± 0.3 N. The NO flux of the 50 wt % nHA-starch-alginate scaffolds was found to be 0.50 ± 0.06 × 10-10 mol/min/mg initially which reduced to 0.23 ± 0.02 × 10-10 over a 24 h period under physiological conditions. As a result, a 99.76% ± 0.33% reduction in a gram-positive bacterium, Staphylococcus aureus and a 99.80% ± 0.62% reduction in the adhered viable colonies of gram-negative bacterium, Pseudomonas aeruginosa were observed, which is a significant stride in the field of antibacterial natural polymers. The surface morphology and pore size were observed to be appropriate for the potential bone cell growth. The material showed no toxic response toward mouse fibroblast cells. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1068-1078, 2019.


Asunto(s)
Antibacterianos , Huesos , Pseudomonas aeruginosa/crecimiento & desarrollo , S-Nitroso-N-Acetilpenicilamina , Staphylococcus aureus/crecimiento & desarrollo , Ingeniería de Tejidos , Andamios del Tejido/química , Células 3T3 , Animales , Antibacterianos/química , Antibacterianos/farmacología , Quitosano/química , Quitosano/farmacología , Ratones , S-Nitroso-N-Acetilpenicilamina/química , S-Nitroso-N-Acetilpenicilamina/farmacología , Almidón/química , Almidón/farmacología
18.
ACS Appl Mater Interfaces ; 11(4): 4523-4530, 2019 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-30607929

RESUMEN

Two major challenges faced by medical devices are thrombus formation and infection. In this work, surface-tethered nitric oxide (NO)-releasing molecules are presented as a solution to combat infection and thrombosis. These materials possess a robust NO release capacity lasting ca. 1 month while simultaneously improving the nonfouling nature of the material by preventing platelet, protein, and bacteria adhesion. NO's potent bactericidal function has been implemented by a facile surface covalent attachment method to fabricate a triple-action coating-surface-immobilized S-nitroso- N-acetylpenicillamine (SIM-S). Comparison of NO loading amongst the various branching configurations is shown through the NO release kinetics over time and the cumulative NO release. Biological characterization is performed using in vitro fibrinogen and Staphylococcus aureus assays. The material with the highest NO release, SIM-S2, is also able to reduce protein adhesion by 65.8 ± 8.9% when compared to unmodified silicone. SIM-S2 demonstrates a 99.99% (i.e., ∼4 log) reduction for S. aureus over 24 h. The various functionalized surfaces significantly reduce platelet adhesion in vitro, for both NO-releasing and non-NO-releasing surfaces (up to 89.1 ± 0.9%), demonstrating the nonfouling nature of the surface-immobilized functionalities. The ability of the SIM-S surfaces to retain antifouling properties despite gradual depletion of the bactericidal source, NO, demonstrates its potential use in long-term medical implants.


Asunto(s)
Antibacterianos/química , Antiinfecciosos/química , Inhibidores de Agregación Plaquetaria/química , Antibacterianos/farmacología , Antiinfecciosos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Plaquetas/efectos de los fármacos , Humanos , Óxido Nítrico/química , Inhibidores de Agregación Plaquetaria/farmacología , S-Nitroso-N-Acetilpenicilamina/química , Staphylococcus aureus/efectos de los fármacos , Propiedades de Superficie
19.
ACS Appl Bio Mater ; 2(6): 2539-2548, 2019 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-33718805

RESUMEN

Devices used for extracorporeal circulation are met with two major medical concerns: thrombosis and infection. A device that allows for anticoagulant-free circulation while reducing risk of infection has yet to be developed. We report the use of a copper nanoparticle (Cu NP) catalyst for the release of nitric oxide (NO) from the endogenous donor S-nitrosoglutathione (GSNO) in a coating applied to commercial Tygon S3™ E-3603 poly(vinyl chloride) tubing in order to reduce adhered bacterial viability and the occurrence thrombosis for the first time in an animal model. Cu GSNO coated material demonstrated a nitric oxide (NO) release flux ranging from an initial flux of 6.3 ± 0.9 ×10-10 mol cm-2 min-1 to 7.1 ± 0.4 ×10-10 mol cm-2 min-1 after 4 h of release, while GSNO loops without Cu NPs only ranged from an initial flux of 1.1 ± 0.2 ×10-10 mol cm-2 min-1 to 2.3 ± 0.2 ×10-10 mol cm-2 min-1 after 4 h of release, indicating that the addition of Cu NPs can increase NO flux up to five times in the same 4 h period. Additionally, a 3-log reduction in S. aureus and 1-log reduction in P. aeruginosa was observed in viable bacterial adhesion over a 24 h period compared to control loops. A Cell Counting Kit-8 (CCK-8) assay was used to validate no overall cytotoxicity towards 3T3 mouse fibroblasts. Finally, extracorporeal circuits were coated and exposed to 4 h of blood flow under an in vivo rabbit model. The Cu GSNO combination was successful in maintaining 89.3% of baseline platelet counts, while the control loops were able to maintain 67.6% of the baseline. These results suggest that the combination of Cu NPs with GSNO increases hemocompatibility and antimicrobial properties of ECC loops without any cytotoxic effects towards mammalian cells.

20.
ACS Biomater Sci Eng ; 5(8): 4002-4012, 2019 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-33443422

RESUMEN

Biological processes such as infection, angiogenesis, and fibroblast proliferation and migration need to be regulated for effective healing of a wound. Failing to do so can delay the overall wound healing and add to the suffering and healthcare cost. Endogenous nitric oxide (NO) is a well-known gasotransmitter in the natural healing process in humans and other mammals. To utilize its inherent ability in the current study, an exogenous NO donor (S-nitroso-glutathione, GSNO) was integrated into a hybrid formulation consisting of a natural polymer (alginate) and a synthetic polymer (poly(vinyl alcohol) (PVA)). The alginate-PVA-GSNO dressings showed a sustained NO release for 72 h that resulted in 99.89 ± 0.40% and 98.93 ± 0.69% eradication of Staphylococcus aureus and Pseudomonas aeruginosa, respectively, which are among the most common causal agents of wound infections. The designed dressings resulted in a 3-fold increase in the proliferation of human endothelial cells when compared with control without GSNO showing its angiogenic potential. In addition, mouse fibroblast cells exposed to leachates from alginate-PVA-GSNO dressings showed significantly higher proliferation when compared to control alginate-PVA showing the NO release from exogenous GSNO in fibroblast proliferation. Fibroblast migration was shown to be much faster with GSNO-based dressings when compared to corresponding control dressings resulting in complete closure of an in vitro wound model within 48 h. The porous dressings also possessed important physical properties such as swelling, water vapor transmission, and moisture content that are desirable for effective wound healing. Overall, this study supports the possibility of using therapeutic alginate-PVA-GSNO dressing to provide a supportive environment for accelerated wound healing.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA