Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Mater Chem B ; 12(18): 4489-4501, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38644661

RESUMEN

Orthopedic device-related infection (ODRI) poses a significant threat to patients with titanium-based implants. The challenge lies in developing antibacterial surfaces that preserve the bulk mechanical properties of titanium implants while exhibiting characteristics similar to bone tissue. In response, we present a two-step approach: silver nanoparticle (AgNP) coating followed by selective laser-assisted surface alloying on commonly used titanium alumina vanadium (TiAl6V4) implant surfaces. This process imparts antibacterial properties without compromising the bulk mechanical characteristics of the titanium alloy. Systematic optimization of laser beam power (8-40 W) resulted in an optimized surface (32 W) with uniform TiAg alloy formation. This surface displayed a distinctive hierarchical mesoporous textured surface, featuring cauliflower-like nanostructures measuring between 5-10 nm uniformly covering spatial line periods of 25 µm while demonstrating homogenous elemental distribution of silver throughout the laser processed surface. The optimized laser processed surface exhibited prolonged superhydrophilicity (40 days) and antibacterial efficacy (12 days) against Staphylococcus aureus and Escherichia coli. Additionally, there was a significant twofold increase in bone mineralization compared to the pristine Ti6Al4V surface (p < 0.05). Rockwell hardness tests confirmed minimal (<1%) change in bulk mechanical properties compared to the pristine surface. This innovative laser-assisted approach, with its precisely tailored surface morphology, holds promise for providing enduring antibacterial and osteointegration properties, rendering it an optimal choice for modifying load-bearing implant devices without altering material bulk characteristics.


Asunto(s)
Aleaciones , Antibacterianos , Escherichia coli , Rayos Láser , Prótesis e Implantes , Plata , Staphylococcus aureus , Propiedades de Superficie , Titanio , Titanio/química , Titanio/farmacología , Plata/química , Plata/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Staphylococcus aureus/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Aleaciones/química , Aleaciones/farmacología , Animales , Pruebas de Sensibilidad Microbiana , Nanopartículas del Metal/química , Calcificación Fisiológica/efectos de los fármacos
2.
Artículo en Inglés | MEDLINE | ID: mdl-37799507

RESUMEN

Inflammatory bowel disease (IBD) has become alarmingly prevalent in the last two decades affecting 6.8 million people worldwide with a starkly high relapse rate of 40% within 1 year of remission. Existing visual endoscopy techniques rely on subjective assessment of images that are error-prone and insufficient indicators of early-stage IBD, rendering them unsuitable for frequent and quantitative monitoring of gastrointestinal health necessary for detecting regular relapses in IBD patients. To address these limitations, we have implemented a miniaturized smart capsule (2.2 cm × 11 mm) that allows monitoring reactive oxygen species (ROS) levels as a biomarker of inflammation for quantitative and frequent profiling of inflammatory lesions throughout the gastrointestinal tract. The capsule is composed of a pH and oxidation reduction potential (ORP) sensor to track the capsule's location and ROS levels throughout the gastrointestinal tract, respectively, and an optimized electronic interface for wireless sensing and data communication. The designed sensors provided a linear and stable performance within the physiologically relevant range of the GI tract (pH: 1-8 and ORP: -500 to +500 mV). Additionally, systematic design optimization of the wireless interface electronics offered an efficient sampling rate of 10 ms for long-running measurements up to 48 h for a complete evaluation of the entire gastrointestinal tract. As a proof-of-concept, the capsule the capsule's performance in detecting inflammation risks was validated by conducting tests on in vitro cell culture conditions, simulating healthy and inflamed gut-like environments. The capsule presented here achieves a new milestone in addressing the emerging need for smart ingestible electronics for better diagnosis and treatment of digestive diseases.

3.
ACS Sens ; 7(4): 960-971, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35333058

RESUMEN

During the γ-radiation sterilization process, the levels of radiation exposure to a medical device must be carefully monitored to achieve the required sterilization without causing deleterious effects on its intended physical and chemical properties. To address this issue, here we have demonstrated the development of an all-printed disposable low-cost sensor that exploits the change in electrical impedance of a semi-interpenetrating polymer network (SIPN) composed of poly(vinyl alcohol) (PVA) and poly(3,4-ethylenedioxythiophene):polystyrenesulfonate (PEDOT:PSS) as a functional polymer composite for radiation sterilization monitoring applications. Specifically, the PEDOT:PSS acts as the electrically conductive medium, while the PVA provides the ductility and stability of the printed sensors. During irradiation exposure, chain scission and cross-linking events occur concurrently in the PEDOT:PSS and PVA polymer chains, respectively. The concurrent scissoring of the PEDOT polymer and cross-linking of the PVA polymer network leads to the formation of a stable SIPN with reduced electrical conductivity, which was verified through FTIR, Raman, and TGA analysis. Systematic studies of different ratios of PEDOT:PSS and PVA mixtures were tested to identify the optimal ratio that provided the highest radiation sensitivity and stability performance. The results showed that PEDOT:PSS/PVA composites with 10 wt % PVA produced sensors with relative impedance changes of 30% after 25 kGy and up to 370% after 53 kGy (which are two of the most commonly used radiation exposure levels for sterilization applications). This composition showed high electrical impedance stability with less than ±5% change over 18 days after irradiation exposure. These findings demonstrate the feasibility of utilizing a printing technology for scalable manufacturing of low-cost, flexible radiation sensors for more effective monitoring of radiation sterilization processes.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes , Polímeros , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Conductividad Eléctrica , Polímeros/química , Esterilización
4.
ACS Appl Mater Interfaces ; 14(7): 9697-9710, 2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35142483

RESUMEN

Many commercially available pH sensors are fabricated with a glass membrane as the sensing component because of several advantages of glass-based electrodes such as versatility, high accuracy, and excellent stability in various conditions. However, because of their bulkiness and poor mechanical properties, conventional glass-based sensors are not ideal for wearable or flexible applications. Here, we report for the first time the fabrication of a flexible glass-based pH sensor suitable for biomedical and environmental applications where flexibility and stability of the sensor are critical for long-term and real-time monitoring. The sensor was fabricated via a simple and facile approach using the cold atmospheric plasma technique in which a pH sensitive silica coating was deposited from a siloxane precursor onto a carbon electrode. In order to increase the sensitivity and stability of the sensor, we employed a postprocessing step which involves annealing of the silica coated electrode at elevated temperatures. This process was optimized to ensure that the crucial properties such as porosity and hydration functionality were balanced to obtain the best and most reliable sensitivity of the sensor. Our sensitivity test results indicated that these sensors exhibit excellent and stable sensitivity with a slope of about 48 mV/pH (r2 = 0.998) and selectivity across a pH range of 4 to 10 in the presence of various cations. The optimized sensor has shown stable sensitivity for a long period of time (30 h of immersion) and in different bending conditions. We demonstrate in this investigation that this flexible cost-effective pH sensor can withstand the sterilization process resulting from ultraviolet radiation and shows repeatable sensitivity with less than ±5 mV potential drift from the sensitivity values of the standard optimized sensor.

5.
Sci Rep ; 12(1): 13927, 2022 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-35977975

RESUMEN

The problematic combination of a rising prevalence of skin and soft tissue infections and the growing rate of life-threatening antibiotic resistant infections presents an urgent, unmet need for the healthcare industry. These evolutionary resistances originate from mutations in the bacterial cell walls which prevent effective diffusion of antibiotics. Gram-negative bacteria are of special consideration due to the natural resistance to many common antibiotics due to the unique bilayer structure of the cell wall. The system developed here provides one solution to this problem through a wearable therapy that delivers and utilizes gaseous ozone as an adjunct therapy with topical antibiotics through a novel dressing with drug-eluting nanofibers (NFs). This technology drastically increases the sensitivity of Gram-negative bacteria to common antibiotics by using oxidative ozone to bypass resistances created by the bacterial cell wall. To enable simple and effective application of adjunct therapy, ozone delivery and topical antibiotics have been integrated into a single application patch. The drug delivery NFs are generated via electrospinning in a fast-dissolve PVA mat without inducing decreasing gas permeability of the dressing. A systematic study found ozone generation at 4 mg/h provided optimal ozone levels for high antimicrobial performance with minimal cytotoxicity. This ozone treatment was used with adjunct therapy delivered by the system in vitro. Results showed complete eradication of Gram-negative bacteria with ozone and antibiotics typically used only for Gram-positive bacteria, which showed the strength of ozone as an enabling adjunct treatment option to sensitize bacteria strains to otherwise ineffective antibiotics. Furthermore, the treatment is shown through biocompatibility testing to exhibit no cytotoxic effect on human fibroblast cells.


Asunto(s)
Infecciones por Bacterias Gramnegativas , Ozono , Dispositivos Electrónicos Vestibles , Antibacterianos/química , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacterias Gramnegativas , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Pruebas de Sensibilidad Microbiana , Ozono/farmacología , Ozono/uso terapéutico
6.
ACS Appl Mater Interfaces ; 14(40): 45752-45764, 2022 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-36173396

RESUMEN

Contamination of meat with pathogenic microorganisms can cause severe illnesses and food waste, which has significant negative impacts on both general health and the economy. In many cases, the expiration date is not a good indicator of meat freshness as there is a high risk of contamination during handling throughout the supply chain. Many biomarkers, including color, odor, pH, temperature, and volatile compounds, are used to determine spoilage. Among these, pH presents a simple and effective biomarker directly linked to the overgrowth of bacteria and degradation of the meat tissue. Low-cost methods for wireless pH monitoring are crucial in detecting spoilage on a large commercial scale. Existing technologies are often limited to short-range detection, with the use of batteries and different electronic components that increases both the manufacturing complexity and cost of the final device. To address these shortcomings, we have developed a cost-effective wireless pH sensor, which uses passive resonant frequency (RF) sensing, combined with a pH-responsive polymer that can be placed within packaged meat products and provide a remote assessment of the risk of microbial spoilage throughout the supply chain. The sensor tag consists of a sensing resonator coated with a pH-sensitive material and a passivated reference resonator operating in a differential frequency configuration. Upon exposure to elevated pH levels >6.8, the coating on the sensing resonator dissolves, which in turn results in a distinct change in the resonant frequency with respect to the reference resonator. Systematic theoretical and experimental results at different pH levels demonstrated that a 20% shift in resonant frequency demarcates the point for spoilage detection. As a proof of concept, the performance of the sensor in remotely detecting the risk of food spoilage was validated in packaged poultry over 10 days. The sensor fabrication process takes advantage of recent developments in the scalable manufacturing of flexible, low-cost devices, including selective laser etching of metalized plastic films and doctor-blade coating of stimuli-responsive polymer films. Furthermore, the biocompatibility of all the materials used in the sensor was confirmed with human intestinal cells (HCT-8 cells).


Asunto(s)
Productos de la Carne , Eliminación de Residuos , Polímeros de Estímulo Receptivo , Humanos , Concentración de Iones de Hidrógeno , Plásticos , Polímeros/química
7.
Acta Biomater ; 154: 83-96, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36162763

RESUMEN

The gastrointestinal (GI) tract, particularly the colon region, holds a highly diverse microbial community that plays an important role in the metabolism, physiology, nutrition, and immune function of the host body. Accumulating evidence has revealed that alteration in these microbial communities is the pivotal step in developing various metabolic diseases, including obesity, inflammatory bowel disease (IBD), and colorectal cancer. However, there is still a lack of clear understanding of the interrelationship between microbiota and diet as well as the effectiveness of chemoprevention strategies, including pre and probiotic agents in modifying the colonic microbiota and preventing digestive diseases. Existing methods for assessing these microbiota-diet interactions are often based on samples collected from the feces or endoscopy techniques which are incapable of providing information on spatial variations of the gut microbiota or are considered invasive procedures. To address this need, here we have developed an electronic-free smart capsule that enables site-specific sampling of the gut microbiome within the proximal colon region of the GI tract. The 3D printed device houses a superabsorbent hydrogel bonded onto a flexible polydimethylsiloxane (PDMS) disk that serves as a milieu to collect the fluid in the gut lumen and its microbiome by rapid swelling and providing the necessary mechanical actuation to close the capsule after the sampling is completed. The targeted colonic sampling is achieved by coating the sampling aperture on the capsule with a double-layer pH-sensitive enteric coating, which delays fluid in the lumen from entering the capsule until it reaches the proximal colon of the GI tract. To identify the appropriate pH-responsive double-layer coating and processing condition, a series of systematic dissolution characterizations in different pH conditions that mimicked the GI tract was conducted. The effective targeted microbial sampling performance and preservation of the smart capsule with the optimized design were validated using both realistic in vitro GI tract models with mixed bacteria cultures and in vivo with pigs as an animal model. The results from 16s rRNA and WideSeq analysis in both in vitro and in vivo studies showed that the bacterial population sampled within the retrieved capsule closely matched the bacterial population within the targeted sampling region (proximal colon). Herein, it is envisioned that such smart sampling capsule technology will provide new avenues for gastroenterological research and clinical applications, including diet-host-microbiome relationships, focused on human GI function and health. STATEMENT OF SIGNIFICANCE: The colonic microbiota plays a major role in the etiology of numerous diseases. Extensive efforts have been conducted to monitor the gut microbiome using sequencing technologies based on samples collected from feces or mucosal biopsies that are typically obtained by colonoscopy. Despite the simplicity of fecal sampling procedures, they are incapable of preserving spatial and temporal information about the bacteria through the gastrointestinal (GI) tract. In contrast, colonoscopy is an invasive and impractical approach to frequently assess the effect of dietary and therapeutic intake on the microbiome and their impact on the health of the patient. Here, we developed a non-invasive capsule that enables targeted sampling from the ascending colon, thereby providing crucial information for disease prediction and monitoring.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Porcinos , Animales , ARN Ribosómico 16S/genética , Microbioma Gastrointestinal/fisiología , Colon , Heces/microbiología , Bacterias
8.
Lab Chip ; 22(1): 57-70, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34826326

RESUMEN

Although serum and fecal biomarkers (e.g., lactoferrin, and calprotectin) have been used in management and distinction between inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS), none are proven to be a differential diagnostic tool between Crohn's disease (CD) and ulcerative colitis (UC). The main challenge with laboratory-based biomarkers in the stool test is the inability to indicate the location of the disease/inflammation in the gastrointestinal (GI) tract due to the homogenous nature of the collected fecal sample. For the first time, we have designed and developed a battery-free smart capsule that will allow targeted sampling of inflammatory biomarkers inside the gut lumen of the small intestine. The capsule is designed to provide a simple and non-invasive complementary tool to fecal biomarker analysis to differentiate the type of IBD by pinpointing the site of inflammatory biomarkers secretion (e.g., small or large bowel) throughout the GI tract. The capsule takes advantage of the rapid change from an acidic environment in the stomach to higher pH levels in the small intestine to dissolve a pH-sensitive polymeric coating as a means to activate the sampling process of the capsule within the small intestine. A swelling polyacrylamide hydrogel is placed inside the capsule as a milieu to collect the sampled GI fluid while also providing the required mechanical actuation to close the capsule once the sampling is completed. The hydrogel component along with the collected GI fluid can be easily obtained from the capsule through the screw-cap design for further extraction and analysis. As a proof of concept, the capsule's performance in sampling and extraction of bovine serum albumin (BSA) and calprotectin - a key biomarker of inflammation - was assessed within the physiologically relevant ranges. The ratio of extracted biomarkers relative to that in the initial sampling environment remained constant (∼3%) and independent of the sampling matrix in both in vitro and ex vivo studies. It is believed that the demonstrated technology will provide immediate impact in more effective IBD type differential diagnostic and treatment strategies by providing a non-invasive assessment of inflammation biomarkers profile throughout the digestive tract.


Asunto(s)
Colitis Ulcerosa , Enfermedad de Crohn , Enfermedades Inflamatorias del Intestino , Biomarcadores , Humanos , Enfermedades Inflamatorias del Intestino/diagnóstico , Intestino Delgado , Complejo de Antígeno L1 de Leucocito
9.
ACS Appl Bio Mater ; 4(7): 5405-5415, 2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35006756

RESUMEN

Chronic nonhealing wounds are a growing socioeconomic problem that affects more than 6 million people annually solely in the United States. These wounds are colonized by bacteria that often develop into biofilms that act as a physical and chemical barrier to therapeutics and tissue oxygenation leading to chronic inflammation and tissue hypoxia. Although wound debridement and vigorous mechanical abrasion techniques are often used by clinical professionals to manage and remove biofilms from wound surfaces, such methods are highly nonselective and painful. In this study, we have developed a flexible polymer composite microneedle array that can overcome the physicochemical barriers (i.e., bacterial biofilm) present in chronic nonhealing wounds and codeliver oxygen and bactericidal agents. The polymeric microneedles are made by using a facile UV polymerization process of polyvinylpyrrolidone and calcium peroxide onto a flexible polyethylene terephthalate substrate for conformable attachment onto different locations of the human body surface. The microneedles effectively elevate the oxygen levels from 8 to 12 ppm once dissolved over the course of 2 h while also providing strong bactericidal effects on both liquid and biofilm bacteria cultures of both Gram-positive (Staphylococcus aureus) and Gram-negative (Pseudomonas aeruginosa) bacterial strains commonly found in dermal wounds. Furthermore, the results from the ex vivo assay on a porcine wound model indicated successful insertion of the microneedles into the tissue while also providing effective bactericidal properties against both Gram-positive and Gram-negative within the complex tissue matrix. Additionally, the microneedles demonstrate high levels of cytocompatibility with less than 10% of apoptosis throughout 6 days of continuous exposure to human dermal fibroblast cells. The demonstrated flexible microneedle array can provide a better approach for increasing the effectiveness of topical tissue oxygenation as well as the treatment of infected wounds with intrinsically antibiotic resistant biofilms.


Asunto(s)
Biopelículas , Infección de Heridas , Animales , Antibacterianos/farmacología , Bacterias , Humanos , Oxígeno/farmacología , Pseudomonas aeruginosa , Staphylococcus aureus , Porcinos , Infección de Heridas/tratamiento farmacológico
10.
RSC Adv ; 10(28): 16313-16322, 2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-35498852

RESUMEN

Gut microbiota plays an important role in host physiology such as obesity, diabetes, and various neurological diseases. Thus, microbiome sampling is a fundamental approach towards better understanding of possible diseases. However, conventional sampling methods, such as endoscopies or colonoscopies, are invasive and cannot reach the entire small intestine. To address this need, a battery-less 3D-printed sampling capsule, which can collect microbiome samples throughout the entirety of the GI tract was designed. The capsule (9 mm × 15 mm) consists of a 3D printed acrylic housing, a fast-absorbing hydrogel, and a flexible PDMS membrane. Fluids containing samples of the microbial flora within the GI tract enter the device through a sampling aperture on the cap of the device. Once the microbiome enters the housing, the hydrogel absorbs the fluid and swells, effectively protecting the samples within its polymeric matrix, while also pushing on the flexible PDMS membrane to block the sampling aperture from further fluid exchange. The retrieved capsule can be readily disassembled due to the screw-cap design of the capsule and the hydrogel can be removed for further bacterial culture and analysis. As a proof of concept, the capsule's bacterial sampling efficiency and the ability to host microbial samples within the hydrogel in a sealed capsule were validated using a liquid culture containing Escherichia coli. The demonstrated technology provides a promising inexpensive tool for direct sampling and assessment of microbes throughout the GI tract and can enable new insights into the role of diet in mediating host-microbe interactions and metabolism.

11.
ACS Appl Bio Mater ; 3(7): 4012-4024, 2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35025476

RESUMEN

We are introducing a wireless and passive strain sensing scheme that utilizes ultrasound imaging of a highly stretchable hydrogel embedded with zinc oxide (ZnO) nanoparticles, named "ZnO-gel". The incorporation of ZnO nanoparticles into a polymer network of the hydrogel improves both its elasticity and strength. It also serves as an ideal biocompatible ultrasound contrast agent that allows remote interrogation of the changes in volume or dimensions of the hydrogel in response to mechanical strains through simple ultrasound imaging. A systematic study of various ratios of ZnO nanoparticle fillers (ranging from 0 to 40% w/w), cross-linked within the poly (DMA-co-MAA) hydrogel, was performed to identify the appropriate ZnO-to-gel ratio that provided the optimal mechanical and ultrasound imaging properties. The results of these investigations showed that 10% w/w of ZnO nanoparticles provided the highest stretchability of 260% with the effective amount of contrast agents to achieve clear visibility of the hydrogel dimension during ultrasound imaging. In general, the applied strain deforms the ZnO-gel specimens by reducing the cross-sectional area at a linear rate of 0.24% area change per % of applied strain for strain levels of up to 250%. Biocompatibility tests with stromal cells (fibroblasts) did not show any acute toxicity of the hydrogel and the ZnO nanoparticles used in this technology. It is anticipated that this technology can be applied to a broad range of wireless and passive monitoring of physiological functions for which microenvironmental strain matters throughout the body, simply by tuning both the mechanical properties of the hydrogel and ZnO nanoparticle concentration.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA