Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.521
Filter
1.
Nutrients ; 16(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732589

ABSTRACT

Sweat rate and electrolyte losses have a large inter-individual variability. A personalized approach to hydration can overcome this issue to meet an individual's needs. This study aimed to investigate the effects of a personalized hydration strategy (PHS) on fluid balance and intermittent exercise performance. Twelve participants conducted 11 laboratory visits including a VO2max test and two 5-day trial arms under normothermic (NOR) or hyperthermic (HYP) environmental conditions. Each arm began with three days of familiarization exercise followed by two random exercise trials with either a PHS or a control (CON). Then, participants crossed over to the second arm for: NOR+PHS, NOR+CON, HYP+PHS, or HYP+CON. The PHS was prescribed according to the participants' fluid and sweat sodium losses. CON drank ad libitum of commercially-available electrolyte solution. Exercise trials consisted of two phases: (1) 45 min constant workload; (2) high-intensity intermittent exercise (HIIT) until exhaustion. Fluids were only provided in phase 1. PHS had a significantly greater fluid intake (HYP+PHS: 831.7 ± 166.4 g; NOR+PHS: 734.2 ± 144.9 g) compared to CON (HYP+CON: 369.8 ± 221.7 g; NOR+CON: 272.3 ± 143.0 g), regardless of environmental conditions (p < 0.001). HYP+CON produced the lowest sweat sodium concentration (56.2 ± 9.0 mmol/L) compared to other trials (p < 0.001). HYP+PHS had a slower elevated thirst perception and a longer HIIT (765 ± 452 s) compared to HYP+CON (548 ± 283 s, p = 0.04). Thus, PHS reinforces fluid intake and successfully optimizes hydration status, regardless of environmental conditions. PHS may be or is an important factor in preventing negative physiological consequences during high-intensity exercise in the heat.


Subject(s)
Exercise , Hot Temperature , Water-Electrolyte Balance , Humans , Water-Electrolyte Balance/physiology , Male , Exercise/physiology , Adult , Young Adult , Female , Sweating/physiology , Dehydration/prevention & control , Dehydration/therapy , Drinking/physiology , Sweat/chemistry , Cross-Over Studies
2.
Biosensors (Basel) ; 14(4)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38667180

ABSTRACT

A lactic acid (LA) monitoring system aimed at sweat monitoring was fabricated and tested. The sweat LA monitoring system uses a continuous flow of phosphate buffer saline, instead of chambers or cells, for collecting and storing sweat fluid excreted at the skin surface. To facilitate the use of the sweat LA monitoring system by subjects when exercising, the fluid control system, including the sweat sampling device, was designed to be unaffected by body movements or muscle deformation. An advantage of our system is that the skin surface condition is constantly refreshed by continuous flow. A real sample test was carried out during stationary bike exercise, which showed that LA secretion increased by approximately 10 µg/cm2/min compared to the baseline levels before exercise. The LA levels recovered to baseline levels after exercise due to the effect of continuous flow. This indicates that the wristwatch sweat LA monitor has the potential to enable a detailed understanding of the LA distribution at the skin surface.


Subject(s)
Lactic Acid , Sweat , Humans , Sweat/chemistry , Lactic Acid/analysis , Monitoring, Physiologic , Wearable Electronic Devices , Biosensing Techniques , Exercise , Skin
3.
Biosens Bioelectron ; 257: 116299, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38636318

ABSTRACT

Skin-interfaced microfluidic patch has become a reliable device for sweat collection and analysis. However, the intractable problems of emptying the microchannel for reuse, and the channel's volumetric capacity limited by the size of the patch, directly hinder the practical application of sweat sensors. Herein, we report an adaptively resettable microfluidic sweat patch (Art-Sweat patch) capable of continuously monitoring both sweat rate (0.2-4.0 µL min-1) and total ionic charge concentration (10-200 mmol L-1). We develop a platform with a vertical and horizontal microchannel combined strategy, enabling repeatedly filling sweat and emptying the microchannel for autonomously resetting and detecting. The variation in the emptied volume is designed to be adaptively identified by the sensor, resulting in enhanced stability and an enlarged volumetric capacity of over 300 µL. By integrating with self-designed wireless transmission modules, the proposed Art-Sweat patch shows product-level wearability and high performance in monitoring variations in regional sweat rate and concentration for hydration status assessment.


Subject(s)
Biosensing Techniques , Electrolytes , Sweat , Sweat/chemistry , Humans , Biosensing Techniques/instrumentation , Electrolytes/chemistry , Wearable Electronic Devices , Equipment Design , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation
4.
ACS Sens ; 9(4): 2075-2082, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38557006

ABSTRACT

Wearable sweat sensors have achieved rapid development since they hold great potential in personalized health monitoring. However, a typical difficulty in practical processes is the control of working conditions for biorecognition elements, e.g., pH level and ionic strength in sweat may decrease the affinity between analytes and recognition elements. Here, we developed a wearable sensing device for cortisol detection in sweat using an aptamer as the recognition element. The device integrated functions of sweat collection, reagent prestorage, and signal conversion. Especially, the components of prestored reagents were optimized according to the inherent characteristics of sweat samples and electrodes, which allowed us to keep optimal conditions for aptamers. The sweat samples were transferred from the inlet of the device to the reagent prestored chamber, and the dry preserved reagents were rehydrated with sweat and then arrived at the aptamer-modified electrodes. Sweat samples of volunteers were analyzed by the wearable sensing device, and the results showed a good correlation with those of the ELISA kit. We believe that this convenient and reliable wearable sensing device has significant potential in self-health monitoring.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Hydrocortisone , Sweat , Wearable Electronic Devices , Sweat/chemistry , Hydrocortisone/analysis , Humans , Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Electrodes , Electrochemical Techniques/instrumentation , Electrochemical Techniques/methods , Indicators and Reagents/chemistry
5.
ACS Appl Mater Interfaces ; 16(15): 19605-19614, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38568178

ABSTRACT

Wearable sweat sensors have received considerable attention due to their great potential for noninvasive continuous monitoring of an individual's health status applications. However, the low secretion rate and fast evaporation of sweat pose challenges in collecting sweat from sedentary individuals for noninvasive analysis of body physiology. Here, we demonstrate wearable textiles for continuous monitoring of sweat at rest using the combination of a heating element and a microfluidic channel to increase localized skin sweat secretion rates and combat sweat evaporation, enabling accurate and stable monitoring of trace amounts of sweat. The Janus sensing yarns with a glucose sensing sensitivity of 36.57 mA cm-2 mM-1 are embroidered into the superhydrophobic heated textile to collect sweat directionally, resulting in improved sweat collection efficiency of up to 96 and 75% retention. The device also maintains a highly durable sensing performance, even in dynamic deformation, recycling, and washing. The microfluidic sensing textile can be further designed into a wireless sensing system that enables sedentary-compatible sweat analysis for the continuous, real-time monitoring of body glucose levels at rest.


Subject(s)
Biosensing Techniques , Wearable Electronic Devices , Humans , Sweat/chemistry , Microfluidics , Glucose/analysis , Monitoring, Physiologic , Textiles , Biosensing Techniques/methods
6.
Int J Biol Macromol ; 267(Pt 2): 131658, 2024 May.
Article in English | MEDLINE | ID: mdl-38636759

ABSTRACT

Moisture evaporation plays a crucial role in thermal management of human body, particularly in perspiration process. However, current fabrics aim for sweat removal and takes little account of basic thermo-regulation of sweat, resulted in their limited evaporation capacity and heat dissipation at moderate/intense scenarios. In this study, a hygroscopic cooling (h-cool) fabric based on multi-functional design, for personal perspiration management, was described. By using economic and effective weaving technology, directional moisture transport routes and heat conductive pathways were incorporated in the construct. The resultant fabric showed 10 times greater one-way transport index higher than cotton, Dri-FIT and Coolswitch fabrics, which contributed to highly enhanced evaporation ability (∼4.5 times than cotton), not merely liquid diffusion. As a result, h-cool fabric performed 2.1-4.2 °C cooling efficacy with significantly reduced sweat consuming than cotton, Dri-FIT and Coolswitch fabrics in the artificial sweating skin. Finally, the practical applications by actually wearing h-cool fabric showed great evaporative-cooling efficacy during different physical activities. Owing to the excellent thermo-moisture management ability, we expect the novel concept and construct of h-cool fabric can provide promising strategy for developing functional textiles with great "cool" and comfortable "dry" tactile sensation at various daily scenarios.


Subject(s)
Sweat , Textiles , Humans , Sweat/chemistry , Hot Temperature , Wettability , Sweating
7.
ACS Sens ; 9(3): 1515-1524, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38447091

ABSTRACT

Stretchable sweat sensors are promising technology that can acquire biomolecular insights for health and fitness monitoring by intimate integration with the body. However, current sensors often require microfabricated microfluidic channels to control sweat flow during lab-on-body analysis, which makes effective and affordable sweat sampling a significant practical challenge. Here, we present stretchable and sweat-wicking patches that utilize bioinspired smart wettable membranes for the on-demand manipulation of sweat flow. In a scalable process, the membrane is created by stacking hydrophobic elastomer nanofibers onto soft microfoams with predefined two-dimensional superhydrophobic and superhydrophilic patterns. The engineered heterogeneous wettability distribution allows these porous membranes to achieve enhanced extraction and selective collection of sweat in embedded assays. Despite the simplified architecture, the color reactions between sweat and chemical indicators are inhibited from directly contacting the skin to achieve a largely improved operation safety. The sensing patches can simultaneously quantify pH, urea, and calcium in sweat through digital colorimetric analysis with smartphone images. The construction with all compliant materials renders these patches soft and stretchy to achieve conformal attachment to the skin. Successfully analyzing sweat compositions after physical exercises illustrates the practical suitability of these skin-attachable sensors for health tracking and point-of-care diagnosis.


Subject(s)
Colorimetry , Sweat , Sweat/chemistry , Capillary Action , Skin , Biomarkers/analysis
8.
Anal Chem ; 96(12): 4997-5005, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38483157

ABSTRACT

In the realm of clinical practice, the concurrent utilization of anticancer medications can enhance their overall therapeutic efficacy. However, it is crucial to acknowledge that the interactions among these anticancer drugs can potentially yield detrimental consequences on their intended outcomes. Consequently, the assessment of both anticancer potency and potential toxic side effects is greatly refined when multiple anticancer drugs are simultaneously detected and evaluated. Here, we designed a wearable electrochemical aptasensor array for monitoring multiple anticancer drugs in sweat. The integrated sensor array consists of three working electrodes modified with three different aptamers (Apt1, Apt2, and Apt3), a Au counter electrode, and a Ag/AgCl reference electrode. Molecular docking simulations were performed to show the binding affinities between three anticancer drugs and their corresponding aptamers. Various eigenvalues were derived from the square-wave voltammetry electrochemical signals, and these data sets were subjected to rigorous analysis through multivariate data analysis techniques. This analytical approach demonstrated exceptional performance by achieving flawless 100% accuracy in the precise identification of nine anticancer drugs consistently at uniform concentrations. Furthermore, the integrated wearable sensor array exhibited impressive capabilities, correctly recognizing all nine anticancer drugs with 100% accuracy and successfully distinguishing between these drugs in artificial sweat samples. The proposed sensor array presents good stability for 15 days. Flexibility tests showed stable device performance after 500 twisting cycles. This innovative wearable sensing array represents a novel approach for achieving real-time monitoring and precise adjustment of drug dosages. It offers invaluable insights for tailoring the treatment of anticancer drugs to individual patients, predicting both drug efficacy and potential adverse reactions within the field of clinical medicine.


Subject(s)
Biosensing Techniques , Sweat , Humans , Sweat/chemistry , Molecular Docking Simulation , Electrodes , Oligonucleotides/analysis , Electrochemical Techniques
9.
ACS Sens ; 9(3): 1565-1574, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38447101

ABSTRACT

Molecular recognition and sensing can be coupled to interfacial capacitance changes on graphene foam surfaces linked to double layer effects and coupled to enhanced quantum capacitance. 3D graphene foam film electrodes (Gii-Sens; thickness approximately 40 µm; roughness factor approximately 100) immersed in aqueous buffer media exhibit an order of magnitude jump in electrochemical capacitance upon adsorption of a charged molecular receptor based on pyrene-appended boronic acids (here, 4-borono-1-(pyren-2-ylmethyl)pyridin-1-ium bromide, or abbreviated T1). This pyrene-appended pyridinium boronic acid receptor is employed here as a molecular receptor for lactate. In the presence of lactate and at pH 4.0 (after pH optimization), the electrochemical capacitance (determined by impedance spectroscopy) doubles again. Lactic acid binding is expressed with a Hillian binding constant (Klactate = 75 mol-1 dm3 and α = 0.8 in aqueous buffer, Klactate = 460 mol-1 dm3 and α = 0.8 in artificial sweat, and Klactate = 340 mol-1 dm3 and α = 0.65 in human serum). The result is a selective molecular probe response for lactic acid with LoD = 1.3, 1.4, and 1.8 mM in aqueous buffer media (pH 4.0), in artificial sweat (adjusted to pH 4.7), and in human serum (pH adjusted to 4.0), respectively. The role of the pyrene-appended boronic acid is discussed based on the double layer structure and quantum capacitance changes. In the future, this new type of molecular capacitance sensor could provide selective enzyme-free analysis without analyte consumption for a wider range of analytes and complex environments.


Subject(s)
Graphite , Lactic Acid , Humans , Lactic Acid/analysis , Graphite/chemistry , Boronic Acids/chemistry , Sweat/chemistry , Electrodes
10.
Anal Chim Acta ; 1299: 342441, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38499429

ABSTRACT

BACKGROUND: In health assessment and personalized medical services, accurate detection of biological markers such as dopamine (DA) and uric acid (UA) in sweat is crucial for providing valuable physiological information. However, there are challenges in detecting sweat biomarkers due to their low concentrations, variations in sweat yield among individuals, and the need for efficient sweat collection. RESULTS: We synthesized CuNi-MOF@rGO as a high-activity electrocatalyst and investigated its feasibility and electrochemical mechanism for simultaneously detecting low-concentration biomarkers UA and DA. Interaction between the non-coordinating carboxylate group and the sample produces effective separation signals for DA and UA. The wearable biomimetic biosensor has a wide linear range of 1-500 µM, with a detection limit of 9.41 µM and sensitivity of 0.019 µA µM-1 cm-2 for DA, and 10-1000 µM, with a detection limit of 9.09 µM and sensitivity of 0.026 µA µM-1 cm-2 for UA. Thus, our sensor performs excellently in detecting low-concentration biomarkers. To improve sweat collection, we designed a microfluidic-controlled device with hydrophilic modification in the microchannel. Experimental results show optimal ink flow at 2% concentration. Overall, we developed an innovative and highly active electrocatalyst, successfully enabling simultaneous detection of low-concentration biomarkers UA and DA. SIGNIFICANCE: This study provides a strategy for sweat analysis and health monitoring. Moreover, the sensor also showed good performance in detecting real sweat samples. This study has shown great potential in future advances in sweat analysis and health monitoring.


Subject(s)
Biosensing Techniques , Wearable Electronic Devices , Humans , Sweat/chemistry , Dopamine/analysis , Uric Acid/analysis , Biosensing Techniques/methods , Biomarkers , Electrochemical Techniques
11.
ACS Appl Mater Interfaces ; 16(14): 18202-18212, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38551998

ABSTRACT

Textile-based sweat sensors display great potential to enhance wearable comfort and health monitoring; however, their widespread application is severely hindered by the intricate manufacturing process and electrochemical characteristics. To address this challenge, we combined both impregnation coating technology and conjugated electrospinning technology to develop an electro-assisted impregnation core-spinning technology (EAICST), which enables us to simply construct a sheath-core electrochemical sensing yarn (TPFV/CPP yarn) via coating PEDOT:PSS-coated carbon fibers (CPP) with polyurethane (TPU)/polyacrylonitrile (PAN)/poloxamer (F127)/valinomycin as shell. The TPFV/CPP yarn was sewn into the fabric and integrated with a sensor to achieve a detachable feature and efficiently monitor K+ levels in sweat. By introducing EAICST, a speed of 10 m/h can be realized in the continuous preparation of the TPFV/CPP yarn, while the interconnected pores in the yarn sheath enable it to quickly capture and diffuse sweat. Besides, the sensor exhibited excellent sensitivity (54.26 mV/decade), fast response (1.7 s), anti-interference, and long-term stability (5000 s or more). Especially, it also possesses favorable washability and wear resistance properties. Taken together, this study provides a crucial technical foundation for the development of advanced wearable devices designed for sweat analysis.


Subject(s)
Biosensing Techniques , Wearable Electronic Devices , Sweat/chemistry , Polyurethanes/chemistry , Carbon Fiber , Textiles
12.
Talanta ; 273: 125865, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38452593

ABSTRACT

Sweat has excellent potential as one of the sources of non-invasive biomarkers for clinical diagnosis. It is relatively easy to collect and process and may contain different disease-specific markers and drug metabolites, making it ideal for various clinical applications. This article discusses the anatomy of sweat glands and their role in sweat production, as well as the history and development of multiple sweat sample collection and analysis techniques. Another primary focus of this article is the application of sweat detection in clinical disease diagnosis and other life scenarios. Finally, the limitations and prospects of sweat analysis are discussed.


Subject(s)
Biosensing Techniques , Sweat , Sweat/chemistry , Biomarkers/analysis , Biosensing Techniques/methods
13.
Talanta ; 274: 125967, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38537349

ABSTRACT

Sweat is an important biofluid with rich physiological information that can evaluate human health condition. Wearable sweat sensors have received widespread attention in recent years due to the benefits of non-invasive, continuous, and real-time monitoring. Currently, an efficient device integrating sweat collection and detection is still needed. Here, a wearable sweat microfluidic system was fabricated for real-time collection and analysis of sweat. The fabricated microfluidic system consisted of four layers, including a skin adhesive layer, a microfluidic layer, an electrode layer, and a capping layer. The sweat collection rate was around 0.79 µL/min, which demonstrated efficient sweat sampling, storage, and refreshing capabilities. Simultaneous detection of multiple sweat biomarkers was achieved with a screen-printed sweat sensing array, which could realize high-precision detection of Na+, K+, and glucose. Moreover, the sensing array also showed good repeatability and stability, with a relative standard deviation of sensitivity of less than 5%. Additionally, human testing was conducted to demonstrate that this microfluidic system can continuously monitor Na+, K+, and glucose in subjects' sweat during exercise, which showed high potential for non-invasive human health monitoring.


Subject(s)
Sweat , Wearable Electronic Devices , Sweat/chemistry , Humans , Lab-On-A-Chip Devices , Sodium/analysis , Glucose/analysis , Potassium/analysis , Microfluidic Analytical Techniques/instrumentation , Biosensing Techniques/instrumentation , Electrodes , Biomarkers/analysis
14.
Lab Chip ; 24(7): 1996-2004, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38373026

ABSTRACT

For the past few years, sweat analysis for health monitoring has attracted increasing attention benefiting from wearable technology. In related research, the sensitive detection of uric acid (UA) in sweat with complex composition based on surface-enhanced Raman spectroscopy (SERS) for the diagnosis of gout is still a significant challenge. Herein, we report a visualized and intelligent wearable sweat platform for SERS detection of UA in sweat. In this wearable platform, the spiral channel consisted of colorimetric paper with Ag nanowires (AgNWs) that could capture sweat for SERS measurement. With the help of photos from a smartphone, the pH value and volume of sweat could be quantified intelligently based on the image recognition technique. To diagnose gout, SERS spectra of human sweat with UA are collected in this wearable intelligent platform and analyzed by artificial intelligence (AI) algorithms. The results indicate that the artificial neural network (ANN) algorithm exhibits good identification of gout with high accuracy at 97%. Our work demonstrates that SERS-AI in a wearable intelligent sweat platform could be a feasible strategy for diagnosis of gout, which expands research on sweat analysis for comfortable and noninvasive health monitoring.


Subject(s)
Biosensing Techniques , Gout , Wearable Electronic Devices , Humans , Sweat/chemistry , Artificial Intelligence , Gout/diagnosis , Spectrum Analysis, Raman , Biosensing Techniques/methods
15.
ACS Appl Mater Interfaces ; 16(8): 9725-9735, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38378454

ABSTRACT

Potentiometric detection in complex biological fluids enables continuous electrolyte monitoring for personal healthcare; however, the commercialization of ion-selective electrode-based devices has been limited by the rapid loss of potential stability caused by electrode surface inactivation and biofouling. Here, we describe a simple multifunctional hybrid patch incorporating an Au nanoparticle/siloxene-based solid contact (SC) supported by a substrate made of laser-inscribed graphene on poly(dimethylsiloxane) for the noninvasive detection of sweat Na+ and K+. These SC nanocomposites prevent the formation of a water layer during ion-to-electron transfer, preserving 3 and 5 µV/h potential drift for the Na+ and K+ ion-selective electrodes, respectively, after 13 h of exposure. The lamellar structure of the siloxene sheets increases the SC area. In addition, the electroplated Au nanoparticles, which have a large surface area and excellent conductivity, further increased the electric double-layer capacitance at the interface between the ion-selective membranes and solid-state contacts, thus facilitating ion-to-electron transduction and ultimately improving the detection stability of Na+ and K+. Furthermore, the integrated temperature and electrocardiogram sensors in the flexible patch assist in monitoring body temperature and electrocardiogram signals, respectively. Featuring both electrochemical ion-selective and physical sensors, this patch offers immense potential for the self-monitoring of health.


Subject(s)
Graphite , Metal Nanoparticles , Graphite/chemistry , Gold/chemistry , Sweat/chemistry , Metal Nanoparticles/chemistry , Electrocardiography
16.
Anal Chem ; 96(9): 3914-3924, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38387027

ABSTRACT

Wearable, noninvasive sensors enable the continuous monitoring of metabolites in sweat and provide clinical information related to an individual's health and disease states. Uric acid (UA) is a key indicator highly associated with gout, hyperuricaemia, hypertension, kidney disease, and Lesch-Nyhan syndrome. However, the detection of UA levels typically relies on invasive blood tests. Therefore, developing a wearable device for noninvasive monitoring of UA concentrations in sweat could facilitate real-time personalized disease prevention. Here, we introduce 1,3,6,8-pyrene tetrasulfonic acid sodium salt (PyTS) as a bifunctional molecule functionalized with Ti3C2Tx via π-π conjugation to design nonenzymatic wearable sensors for sensitive and selective detection of UA concentration in human sweat. PyTS@Ti3C2Tx provides many oxidation-reduction active groups to enhance the electrocatalytic ability of the UA oxidation reaction. The PyTS@Ti3C2Tx-based electrochemical sensor demonstrates highly sensitive detection of UA in the concentration range of 5 µM-100 µM, exhibiting a lower detection limit of 0.48 µM compared to the uricase-based sensor (0.84 µM). In volunteers, the PyTS@Ti3C2Tx-based wearable sensor is integrated with flexible microfluidic sweat sampling and wireless electronics to enable real-time monitoring of UA levels during aerobic exercise. Simultaneously, it allows for comparison of blood UA levels via a commercial UA analyzer. Herein, this study provides a promising electrocatalyst strategy for nonenzymatic electrochemical UA sensor, enabling noninvasive real-time monitoring of UA levels in human sweat and personalized disease prevention.


Subject(s)
Biosensing Techniques , Nitrites , Transition Elements , Wearable Electronic Devices , Humans , Uric Acid/analysis , Titanium/analysis , Sweat/chemistry
17.
Arch Dis Child ; 109(4): 304-307, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38253430

ABSTRACT

OBJECTIVE: To verify the rate and predictors of 'quantity not sufficient' (QNS) among Brazilian infants younger than 3 months with positive newborn screening (NBS) for cystic fibrosis (CF). DESIGN: Prospective, population-based study. SETTING: Public Statewide Newborn Screening Programme where the incidence rate of CF is ≈1:11 000. PATIENTS: Subjects with positive two-tiered immunoreactive trypsinogen. INTERVENTIONS: Sweat induction and collection were performed in the same facility; one sweat sample was obtained per individual. MAIN OUTCOME MEASURES: The QNS rate and its predictors; analysis corresponded to the day of sweat collection. RESULTS: Among the 975 participants, QNS rates for 10 and 15 µL were 3.6% (95% CI 2.5% to 4.9%) and 8.3% (95% CI 6.6% to 10.2%). Infants weighing >3056 and >3845 g and with gestational age higher than 37 weeks had a greater likelihood (5.5 and 6.7, and 2.7 and 5.8 times more, respectively) of avoiding QNS than their peers. CONCLUSION: QNS rates fulfilled the requirements, but predictors differed from those recommended by the Cystic Fibrosis Foundations guidelines.


Subject(s)
Cystic Fibrosis , Pilocarpine , Infant, Newborn , Infant , Humans , Cystic Fibrosis/diagnosis , Cystic Fibrosis/epidemiology , Iontophoresis , Sweat/chemistry , Prospective Studies , Neonatal Screening , Trypsinogen , Cystic Fibrosis Transmembrane Conductance Regulator , Chlorides/analysis
18.
Lab Chip ; 24(4): 776-786, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38197467

ABSTRACT

Thread-based microfluidic colorimetric sensors have been deemed a potential tool that may be incorporated into textiles for non-invasive sweat analysis. Nevertheless, their poor performance significantly limits their practical uses in sweat glucose detection down to 20 µM. Herein, a microfluidic glucose sensing device containing a salt-responsive thread is developed for the highly sensitive detection of glucose in human sweat. By grafting a zwitterionic polymer brush-which could react to ionic strength by changing the conformation of the polymer chains from the collapsing state to the stretching state-onto the cotton thread, the salt-responsive thread was created. Compared to the pristine cotton thread, the modified thread has better ion-capture capabilities, a more noticeable swelling effect, and a higher ability to absorb water. These enable a significant enrichment of glucose when the saline solution passes through it. The salt-responsive thread was employed to construct a thread/paper-based microfluidic sensing device for the monitoring of glucose in artificial sweat, exhibiting a sensitivity of -0.255 µM-1 and a detection limit of 14.7 µM. In comparison to the pristine cotton thread-based device, the performance is significantly superior. Using a hydrophobic fabric and salt-responsive threads, a glucose-sensing headband was prepared for on-body sweat glucose monitoring. With the use of a smartphone-based image analysis system, the headband can detect the concentration of glucose in a volunteer's perspiration. Using the thread-based salt-responsive zwitterionic polymer brush might offer a novel approach to creating wearable sweat sensors with extremely high sensitivity.


Subject(s)
Biosensing Techniques , Sweat , Humans , Sweat/chemistry , Microfluidics , Blood Glucose Self-Monitoring , Blood Glucose/analysis , Glucose/analysis , Polymers
19.
Clin Chem Lab Med ; 62(6): 1118-1125, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38253354

ABSTRACT

OBJECTIVES: Urea and creatinine concentrations in plasma are used to guide hemodialysis (HD) in patients with end-stage renal disease (ESRD). To support individualized HD treatment in a home situation, there is a clinical need for a non-invasive and continuous alternative to plasma for biomarker monitoring during and between cycles of HD. In this observational study, we therefore established the correlation of urea and creatinine concentrations between sweat, saliva and plasma in a cohort of ESRD patients on HD. METHODS: Forty HD patients were recruited at the Dialysis Department of the Catharina Hospital Eindhoven. Sweat and salivary urea and creatinine concentrations were analyzed at the start and at the end of one HD cycle and compared to the corresponding plasma concentrations. RESULTS: A decrease of urea concentrations during HD was observed in sweat, from 27.86 mmol/L to 12.60 mmol/L, and saliva, from 24.70 mmol/L to 5.64 mmol/L. Urea concentrations in sweat and saliva strongly correlated with the concentrations in plasma (ρ 0.92 [p<0.001] and 0.94 [p<0.001], respectively). Creatinine concentrations also decreased in sweat from 43.39 µmol/L to 19.69 µmol/L, and saliva, from 59.00 µmol/L to 13.70 µmol/L. However, for creatinine, correlation coefficients were lower than for urea for both sweat and saliva compared to plasma (ρ: 0.58 [p<0.001] and 0.77 [p<0.001], respectively). CONCLUSIONS: The results illustrate a proof of principle of urea measurements in sweat and saliva to monitor HD adequacy in a non-invasive and continuous manner. Biosensors enabling urea monitoring in sweat or saliva could fill in a clinical need to enable at-home HD for more patients and thereby decrease patient burden.


Subject(s)
Creatinine , Renal Dialysis , Saliva , Sweat , Urea , Humans , Urea/analysis , Urea/blood , Saliva/chemistry , Creatinine/blood , Creatinine/analysis , Sweat/chemistry , Female , Male , Cohort Studies , Middle Aged , Aged , Kidney Failure, Chronic/therapy , Kidney Failure, Chronic/blood , Adult , Biomarkers/analysis , Biomarkers/blood
20.
ACS Nano ; 18(3): 2335-2345, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38189251

ABSTRACT

Stretchable sweat sensors have become a personalized wearable platform for continuous, noninvasive health monitoring through conformal integration with the human body. Typically, these devices are coupled with soft microfluidic systems to control sweat flow during advanced analysis processes. However, the implementation of these soft microfluidic devices is limited by their high fabrication costs and the need for skin adhesives to block natural perspiration. To overcome these limitations, a stretchable and smart wettable patch has been proposed for multiplexed in situ perspiration analysis. The patch includes a porous membrane in the form of a patterned microfoam and a nanofiber layer laminate, which extracts sweat selectively from the skin and directs its continuous flow across the device. The integrated electrochemical sensor array measures multiple biomarkers simultaneously such as pH, K+, and Na+. The soft sensing patch comprises compliant materials and structures that allow deformability of up to 50% strain, which enables a stable and seamless interface with the curvilinear human body. During continuous physical exercise, the device has demonstrated a special operating mode by actively accumulating sweat from the skin for multiplex electrochemical analysis of biomarker profiles. The smart wettable membrane provides an affordable solution to address the sampling challenges of in situ perspiration analysis.


Subject(s)
Biosensing Techniques , Wearable Electronic Devices , Humans , Sweat/chemistry , Skin , Lab-On-A-Chip Devices
SELECTION OF CITATIONS
SEARCH DETAIL
...