Materials, Structure and System Composition of Sweat Sensor: Comparison
Please note this is a comparison between Version 2 by Camila Xu and Version 1 by Hongju Mao.

Sweat, as a biofluid that is easy to extract and contains a variety of biomarkers, can provide various types of physiological information for health monitoring.

  • health monitoring
  • sweat analysis
  • wearable device
  • sensing platform
  • non-invasive detection

1. Sensing Materials

1.1. Common Materials for Electrodes

Common counter-electrode materials include gold, platinum and carbon. The reference electrode is typically Ag/AgCl. The working electrode is divided into enzymatic reaction electrode, molecular selection electrode [115][1] and ion-selective electrode. When the sensitive membrane of the ion selective electrode contacts the solution, an electrical potential is generated at the interface that is directly related to the corresponding ion concentration. For example, the multiplexed sensor proposed by Mei et al. integrates five electrochemical sensors detecting lactate, glucose, pH, Cl and urea, of which the detection of lactate and glucose uses enzyme-modified electrodes, using current changes to detect concentration; pH, Cl and urea are sensed by electrodes modified with PANI, Ag treated with FeCl3 and NH4+ ion-selective membranes, respectively. As mentioned above, sensing materials are also gradually developing in the direction of multifunctional multi-modulus in the form of arrays. For example, the multiplexed sensor proposed by Mei et al. [109][2] integrates five electrochemical sensors, lactate, glucose, pH, Cl and urea, of which the detection of lactate and glucose uses enzyme-modified electrodes that rely on current changes to detect concentration; pH, Cl and urea are sensed by electrodes modified with PANI, Ag treated with FeCl3 and NH4+ ion-selective membranes, respectively. As mentioned above, sensing materials are helping to improve devices in a multifunctional and multimodal way in the form of arrays.

1.2. Nanomaterials

As materials science continues to advance, the application of nanomaterials has become increasingly sophisticated. Due to their unique properties, such as porosity, rigidity, and impedance, as well as their customizable electrical and wetting properties, nanomaterials have been widely used in the fabrication of sensing materials, substrates, and other components to enhance sensor performance. Lin et al. [120][4] utilized N-GODs to anchor a PANI matrix as a sensing electrode, resulting in improved sensitivity and stability while avoiding the cracking commonly associated with rigid electrodes. Furthermore, the modification of electrodes with metal nanomaterials can optimize sensing performance. For example, the modification of a Prussian blue electrode with gold nanoparticles (AuNPs) yielded a larger amperometric response [121][5]. In the fabrication of electrochemical electrodes, metal nanomaterials are often combined with redox graphene (rGO) [122[6][7],123], which serves as a supporting substrate to enhance sensitivity. Due to its high electron mobility and large active surface area, this composite often leads to significant improvements in the sensitivity and accuracy of sensors. Additionally, such nanocomposites generally exhibit excellent electrocatalytic activity [124,125][8][9]. Yu et al. [126][10] developed a CuxO-nanoflake (NFs)/Cu nanoparticles (NPs) nanocomposite as a sensing material. The sensor was enriched with more active sites, a low detection limit, high selectivity, and resistance to bending and twisting. Assaduzzaman et al. [9][11] developed a flexible, multifunctional patch based on laser-induced graphene (LIG) combined with hybrid nano-porous carbon (H-NPC). Sensors based on nanomaterials exhibit excellent electrochemical activity, reliability and a wide detection range.

1.3. Two-Dimensional Materials

Graphene: As a monolayer two-dimensional material, Graphene has long been of interest to researchers. Special variants of graphene, such as graphene oxide and reduced graphene oxide, are widely used in wearable sweat sensors. Their high surface-area-to-volume ratio provides a large number of reaction sites, excellent electrocatalytic performance and high electron mobility. Graphene is often used as a substrate for electrodes and is combined with other materials or modification methods to enhance the overall performance of the electrode. After modification with graphene, the sensing electrode shows high sensitivity, good selectivity and flexibility, enabling the detection of small-molecule concentrations such as glucose [127[12][13],128], lactate [9][11], cortisol [26][14] and uric acid [129][15]. Wang, Y et al. [130][16] synthesized reduced graphene oxide (RGO) in situ on cotton fabric (CF). This imparted softer physical properties to the graphene material while achieving a fast response and excellent selectivity. In addition, the electrodes can be fabricated by engraving graphene films into specific patterns using laser-engraving techniques. Wang, M et al. [43][17] fabricated an electrode based on laser-engraved graphene (LEG) and Molecularly Imprinted Polymers (MIP) that can be repeatedly regenerated in situ. This electrode can be used to assess amino acid intake levels [131][18]. LEG can likewise be modified with nanomaterials (e.g., AuNPs) for highly sensitive electrochemical detection. According to the research conducted by Tu et al. [132][19], the deposition of AuNPs via pulsed potential results in their uniform distribution throughout the mesoporous graphene structure. This enhances their catalytic ability and provides a large number of specific binding sites. Laser-induced graphene (LIG), on the other hand, produces graphene directly on polymers using lasers without needing a film [133][20]. Feng et al. [134][21] demonstrated the use of LIG and single-walled carbon nanotubes (SWCNT) in the development of a sweat sensor. The LIG fabrication process is both rapid, taking less than a minute, and inexpensive. Additionally, laser-scribed graphene (LSG) has also been shown to have potential for use in chemical sensing [135][22]. The emergence of LEG and LIG has made the large-scale fabrication of graphene devices increasingly promising.
MXene: MXene is a novel material discovered by Gogosti in 2011 [136][23]. It is a two-dimensional transition metal carbide and nitride with a tunable structure and can exhibit excellent electrical, optical, catalytic and mechanical properties when appropriately modified [137][24]. The formation of composites with MXene and other materials, such as Pt and polymers, can result in enhanced performance, making it suitable for use in wearable devices. Ti3C2Tx was the first synthesized MXene material due to its low formation energy and in wearable sweat-sensor studies; Ti3C2Tx is a commonly used MXene material [138][25]. Ti3C2Tx exhibits high electrical conductivity, excellent hydrophilicity, and a large number of active sites and terminal functional groups. These properties reduce signal interference and promote the adsorption of gas and water molecules, making Ti3C2Tx well-suited for use in the manufacture of sensors [139][26]. Sharifuzzaman et al. [140][27] reported a catalyst composed of MXene/fluoropolymer-derived porous TiO2 nanomaterials. On the other hand, Magesh et al. [45][28] developed a carbon (Pearlman catalyst) using a MXene/palladium hydroxide-loaded composite (MXene/Pd(OH)2/C) for electrochemical sensors, achieving good selectivity, sensitivity and stability. Li et al. [141][29] used MXene to develop a fully optimized glucose sensor. Pt/MXene nanomaterials were fabricated using Pt nanoparticles for glucose detection on MXene nanosheets, yielding a wide linear range under neutral conditions. The sensor substrate was a conductive hydrogel synthesized from MXene and polyvinyl alcohol (PVA), providing reliable stability to the sensor.

1.4. Hydrogel

Hydrogel-based flexible sensors possess numerous properties such as compatibility, self-healing, and adhesion [142][30]. Due to their water content of up to 90%, they exhibit excellent biocompatibility, electrical conductivity, and flexibility. The use of hydrogel as a sensor substrate or electrode material can improve the sensing performance. The elastic hydrogel system results in a skin-like base that can significantly improve the wearability and mechanical deformability of the device. It has been used in applications such as tissue engineering [143[31][32],144], drug delivery carriers [145][33], soft robots [146[34][35],147], and wound dressings [148,149][36][37]. Wang et al. [150][38] used TPU film to combine hydrogel and conductive material to provide the sensor with excellent mechanical and electrical properties at the same time, verifying the ability of hydrogel to improve comfort.
Gao et al. [151][39] used porous hydrogel instead of liquid electrolyte on sensor electrodes, maintaining better conductivity while achieving improved swelling and biocompatibility. A recent study showed that glycerin gel can also achieve alternative functions while providing anti-bending and sweat absorbing effects [127][12]. As a new type of gel, noble metal aero-/hydrogels possess the properties of both aerogel materials and noble metal nanoparticles [152,153][40][41]. Their highly branched, fused nanowire structure imparts inherent self-healing and flexible properties, making them promising for flexible sensing [154,155][42][43]. Li et al. [156][44] designed a bis-structured Pt-Ni hydrogel that achieved the aforementioned excellent properties and remained stable for two months. Chen et al. [129][15] reported a wearable biosensing platform based on metal aerogels using three-dimensional porous bis-structured aerogels (N-rGO/Au DAs) consisting of gold nanowires and N-doped graphene nanosheets as working electrodes. The high surface area and porous structure, as well as the synergistic effect of Au and N-rGO aerogels, enable better catalytic and electron-transfer properties than mono aerogels, facilitating non-enzymatic uric acid sensors.

1.5. Organic Electrochemical Transistors

An organic electrochemical transistor (OECT) is a low-cost, highly sensitive device that responds to physical stimuli, chemical functionalization, and shape changes. The OECT is composed of an ion-permeable organic semiconductor channel (OSC) connecting the source and drain electrodes. Its operation involves the conversion of the ionic current between the electrodes into a modulation of the polymer film conductivity, allowing for the detection of electrolyte solution concentrations. Poly(3,4-ethylenedioxythiophene):poly(styrenesulfonate) (PEDOT:PSS) is commonly used as a conductive pathway between the source and drain of the OECT due to its ability to amplify signals, ease of miniaturization, and excellent biocompatibility [157,158][45][46]. PEDOT:PSS-based OECTs exhibit high transconductance (≈2 mS) at low voltages (<0.5 V) [159][47], making them promising for use in biosensing applications. However, these OECTs are sensitive to ionic strength [160][48] and the PEDOT:PSS channel accepts all cation insertions from the analyte, resulting in a lack of selectivity [161][49]. The selectivity of OECTs for analytes in sweat can be improved through the use of an ion-selective membrane (ISM), which permits only specific ions to pass through. In 2014, Sessolo et al. [162][50] first proposed the incorporation of an ISM into an OECT for the detection of K+, inspiring subsequent research in the field of wearable sweat sensors. Coppedè et al. [163][51] later proposed an OECT using an ISM for the detection of Ca2+, with the device being fabricated by functionalizing textile fibers. It is worth noting that many recent studies on OECT sweat sensors have incorporated textiles as a platform. Textiles are easy to prepare and can be integrated into clothing, and the detection of target analytes is not limited to ions. Tao et al. [164][52] developed a fabric-based OECT lactate sensor that enables PEDOT:PSS to exhibit high transconductance in both depletion and accumulation modes. Fang et al. [165][53] addressed the issue of insufficient transconductance in fiber OECTs by modifying carbon nanotube (CNT) fiber grids. Qing et al. [166][54] further integrated fiber OECTs with thermoelectric fabrics, using a manufacturing substrate composed of cotton/poly(3,4-ethylenedioxythiophene):poly(styrenesulfonate)/dimethylsulfoxide/(3-glycidyloxypropyl)trimethoxysilane (PDG) yarn, which is lightweight, strong, and sweat-resistant.

1.6. Other Materials

There are numerous reports on fiber-based wearable electronics, with many studies focusing on the application of fibers for physical sensing, such as pressure [167][55] and thermal [168][56] sensing. Instead, Wu et al. [169][57] focused on biochemical parameters to develop a flexible microelectronic fiber that can be woven into textile substrates. Detection of UA oxidation or Na+ concentration can be achieved by modifying ion-sensitive membranes or nanoparticles.
Garcia-Rey et al. [5][58] conducted a proof-of-concept study on alginate-based biological systems to evaluate the efficacy of the colorimetric determination of sweat glucose. Alginate beads were obtained by integrating enzyme assays, consisting of GOx, HRP, and TMB, into the alginate scaffold. The porosity of the beads enabled highly sensitive glucose detection, and a similar system has been used in previous studies to calibrate and detect lactate in sweat [170][59]. Furthermore, another report stated that the integration of TiO2 nanotubes into alginate can provide enhanced hydrophilicity, further improving the sensing performance of this material [171][60]. The touch-based cortisol sensor reported by Tang et al. [116][61] embeds Prussian blue redox probes within a molecularly imprinted polymer (imprinted polypyrrole, PPy, film) network. The selective binding of cortisol to the imprinted PPy film impedes PB charge transfer, resulting in a rapid change in current.
Tang et al. [117][62] reported a WO3-based pH sensor. According to the article, conventional pH sensing methods primarily utilize organic materials, which carry the risk of biological toxicity. As an alternative, WO3 is an inorganic and non-toxic material that is sensitive to hydrogen ions. However, its exchange rate is very low. The embedding of lattice H+ transforms WO3 from a monoclinic phase to a cubic phase (HxWO3), enhancing its H+ exchange capacity and reducing its resistance, thereby improving detection sensitivity and response speed.
The use of composite sensing materials is becoming an increasingly popular trend in the research into wearable sweat sensors. While individual materials may have specific limitations, composite materials can be specifically designed to balance the strengths and weaknesses of different materials to achieve the desired functionality. Nanomaterials are also gradually becoming significant in this field. The various materials mentioned above can be nanosized or combined with nanomaterials to produce interfaces or electrodes with a large number of active sites, a large surface area, and good mechanical properties, thereby optimizing sensor performance.

2. Forms and Structure

2.1. Development of Sweat Sensor Forms

In the early years of wearable sensor research, various forms of sensors, including tattoos, wristbands, and patches, have been reported. In 2012, Professor Joseph Wang’s research team reported a tattoo-based electrochemical sensor [172][63]. Based on this research, many applications, including the detection of ammonium [173][64], lactate [75][65], and sodium [110][66], have been developed in vitro and attached to the skin using transfer technology. Tattoo sensors integrated with carbon fiber exhibit excellent immunity to disturbances such as stretching, bending, and skin abrasion, and have better electrical performance than screen-printed electrodes. In addition, these sensors can be combined with artistic tattoos, which has attracted the attention of many researchers. In practice, tattoo-based sensors are more like a new type of electrode. While research on tattoo-based sensors has seen significant improvements, their popularity in subsequent research has gradually declined. Katseli et al. [7][67] proposed a 3D-printed electrochemical ring for sweat glucose detection. The complex fabrication process of wearable sensors is simplified by embedding conductive plastic electrodes in a ring holder and using a dual-extrusion 3D printer and commercially available filaments in a one-step printing process. In addition to the above research, many sweat sensors are now manufactured in patch form, including readable disposable sensor patches and system-based patches.
Currently, the research focus has gradually shifted, first from single-parameter to multi-parameter detection. The human body system is complex and constantly in a dynamically changing environment. As such, it is difficult to fully express the physiological health status of the human body by interpreting a single parameter. Consequently, more research is now focused on multi-parameter detection, including metabolites and electrolytes [174][68], sweat analytes and sweat rate [175][69], providing data on multiple biomarkers for analysis.
From single-component components to system-level studies, Gao et al. [6,176][70][71] proposed a sensor array capable of multiplexed sweat composition analysis, in 2016. The array was connected to integrated circuits and flexible circuit boards to simultaneously detect sweat metabolites, such as lactic acid and glucose, and electrolyte ions, including pH, calcium, sodium, and potassium. This study aimed to propose a design scheme for wearable sweat sensors from the perspectives of system integration, signal calibration, and multiplexing. This groundbreaking work provided researchers with a more complete system-level design.
Multi-function sweat sensors broaden the information collection range of single-channel sensors, and further expansion (e.g., combining physical signal detection such as acoustics) is expected to provide a more comprehensive picture of human health information. Sempionatto et al. [177][72] report a multiplexed sensor capable of not only detecting multiple biomarkers in sweat, but also integrating an ultrasound transducer to detect blood pressure and heart rate. One of the issues that must be considered when multiplexing integrated sensors is to prevent signal crosstalk from affecting the reliability of the data from each sensor. A solution to the signal crosstalk problem was proposed that spatially separates components at optimal distances and prevents crosstalk between acoustic and electrochemical transducers with solid-state ultrasound and sensing hydrogel layers.
While the concept of multiplexed detection and system design research is on the rise, making sensor systems lighter is also an important issue in the development of complex sensors. Gao et al. [178][73] proposed a lightweight flexible sensor based on their previous research. To achieve this, the researchers aimed to make materials that fit the body lighter, such as paper-based and textile sensors.
A disadvantage of using sweat as a detection target is the small sample size available at rest. To overcome this limitation, flexible sensors with integrated sweat-stimulation capabilities are being developed. Sam et al. [179][74] used iontophoresis electrodes to deliver agonists to stimulate sweat gland secretion, which has gradually become the main method for sweat induction. Research into natural sweat production is also increasing due to its non-invasive nature.
Research on sweat sensors does not stop at simply completing the detection function. Researchers also expect them to have therapeutic capabilities. Lee et al. [128][13] used microneedles to design sweat sensors capable of transcutaneous drug delivery, adding medical value to wearable devices.
As wearable sweat sensors move closer to practical applications, portability and miniaturization are inevitable trends for clinical and everyday consumer markets [180,181][75][76]. One important method for achieving device miniaturization is to replace traditional standard discrete components with integrated circuits. Wang et al. [182][77] reported an ultra-small wearable sweat sensing system with a size of only 1.5 cm × 1.5 cm. Its core processing chip, the MS02 chip, integrates functional circuits such as amplification and filtering, which greatly reduces the overall system area while maintaining sensor performance.

2.2. Structure of Sweat Sensor

Wearable sweat sensors can be roughly divided into two categories based on whether they have a sweat transport structure: patch and microfluidic-based devices. The former undergoes electron transfer or optical reactions directly upon contact with sweat, while the latter collects or transports sweat to electrodes or reagents through microfluidic channels for detection. Taking electrochemical detection as an example, when the detection target is relatively simple, sweat can be brought directly into contact with the electrode to achieve rapid real-time monitoring. However, when multiple analytes are involved, the lack of sweat sample becomes more noticeable, making it difficult to supply multiple electrodes for simultaneous analysis. Timed sampling with an absorbent patch or a regular sweat collector can solve this problem, but it increases the effort required [183][78]. In addition, changes in sweat rate over time and across body parts, combined with factors such as epidermal contaminants and sweat evaporation, can easily cause fluctuations in the results of sweat sensor patches [184][79]. By incorporating microfluidic channels, sweat can be collected and distributed to different sensing electrodes on demand, overcoming problems such as inadequate sweat sample volume, instability, and sweat evaporation. The effects of contamination can also be reduced with specially designed channels.
Microfluidics: Microfluidic systems have the advantages of simple structure, strong ductility, and storage and transport capabilities. By designing the size (e.g., capillary [115][1], width variation), structure (e.g., serpentine structure, fiber, reservoir distribution), and other properties of the microfluidic channels, they can acquire specific capabilities. Examples include designing numerous pathways for the multi-modal analysis of sweat [96][80] and building attractive fluid dynamics and reservoirs for rapid filling [185][81]. Microfluidics have long been used in wearable sweat sensors as channels for the rapid sampling, collection, and analysis of sweat. Microfluidic sensing patches can be mass-produced using techniques like Roll-to-Roll (R2R) printing [186][82], a low-cost rotary screen printing process. Biocompatible and stretchable materials including PDMS [90][83] (polydimethylsiloxane) and PET (polyethylene terephthalate) are commonly used to construct microfluidic channels, allowing sensors to be integrated anywhere on the body [187][84].
As mentioned above, the structure of microfluidic channels can be customized. However, a more complex microfluidic system does not necessarily result in better performance. For instance, the flowmeter-based sweat rate sensor reported by Kwon et al. [46][85] relies on a short, straight microchannel that is thermally coupled to sweat to accurately measure temperature differences and obtain flow values. Microfluidic channels can also be designed as three-dimensional structures, allowing the vertical integration of multiple modules for sampling, transport, sensing, and storage [188][86].
The microfluidic channel is a semi-open system that depends on capillary action to transport sweat during collection. As such, it is important to control the fluid flow within the channel. Shi et al. [118][87] proposed the addition of forward and reverse Tesla valves to the inlet and outlet to prevent inlet backflow, and reduce contact between the outlet and the environment. This design is highly suitable for low-flow-rate conditions, such as sweat monitoring.
With the ongoing advancement of materials science, new microfluidic systems have been developed to address the limitations of traditional hollow microfluidic channels, such as contamination, miniaturization, and diffusion of chemical agents. Currently, there are studies exploring the combination of nanofibers with microfluidic channel materials. Mei et al. [109][2] reported a nanofiber-based microfluidic analysis system (NFMAS). This system utilizes highly porous and hydrophilic polyimide/sodium dodecyl sulfate (PI/SDS) nanofiber films as a microfluidic network substrate for collecting and conducting sweat. The system features both electrochemical and colorimetric sensing modules, which are separated by nanofibers. These sensors exhibit excellent mechanical properties and resistance to contamination. In subsequent research, the same group continued to apply this nanofiber microfluidic technology to fabricate flexible sensors [189][88], with the goal of better improvement.
Another approach is to use paper-based microfluidics. By cutting paper into a channel shape, sweat can be transported by capillary action [190,191][89][90]. Also, paper can be folded to form three-dimensional structures, allowing the creation of more complex channels. Liang et al. [192][91] demonstrated a three-dimensional paper-based microfluidic electrochemical device (3D-PMED). The device is constructed by prefabricating a pattern on cellulose paper and folding it into a five-layer stacked structure containing a sweat collector, vertical channel, transverse channel, electrode layer, and sweat evaporator.

3. System and Module

3.1. Sweat Collection

There are several common methods for collecting sweat using wearable sensing systems. One approach is to use external stimuli, such as iontophoresis, to induce the release of sweat from the skin. Another approach is to collect sweat produced naturally during high-intensity exercise or to absorb sweat from the skin under resting conditions [190][89].
Iontophoresis: Iontophoresis involves the use of a pair of positive and negative electrodes. The anode releases pilocarpine or other long-acting sweat-inducing drugs, such as carbachol [196][92], to induce sweating without significant stimulation. The anode used in iontophoresis is often positioned in the center of the measuring electrodes [197][93] to enable real-time sweat sampling or detection. However, due to the mixing of sweat and gel and the lack of dynamic sweat sampling, this method can result in a limited number of effective samples and reduced measurement accuracy [43][17]. By combining iontophoresis with microfluidics [188][86], induced sweat can be collected and directly transferred to the sensing electrode, resulting in a more stable and quantifiable sample. Bolat et al. [196][92] reported on a specialized wearable, single-step sensing platform that integrates iontophoresis with microfluidics. The iontophoresis electrode is positioned directly around the PDMS microchannel, stimulating sweat glands near the microfluidic inlet. This allows secreted sweat to precisely enter the channel and be delivered to the sensor. A similar approach has been used by Wang et al. [43][17] in their sweat sensing patches.
Hydrophilic/Hydrophobic Substrate: Zhang et al. [97][94] developed a stretchable, colorimetric sweat sensor that utilizes the large wettability gradient between a superhydrophobic substrate and superhydrophilic assays to collect sweat. Superhydrophilic assays are composed of elastomeric nanofiber textiles modified with SiO2 nanoparticles, rather than the more commonly used filter papers. Superhydrophobic substrates are created by modifying the surface of elastomer substrates with nanoparticles. This elastic substrate, featuring a wetting gradient and favorable comfortability, is well-suited for non-invasive and efficient sweat collection.
Hydrogel for Natural Sweat Extraction: Skin-conforming hydrogels have a hydrophilic interface. The hydrogel that adheres directly to the skin has the ability to reduce Laplace pressure, which prevents sweat from forming on the skin surface. In addition, water-based patches are an effective matrix for metabolite extraction, and the proper thickness of hydrogel patches can substantially improve the sensor’s capability to collect biomarkers [76][95]. Lin et al. [76][95] developed a wearable hydrogel patch that leverages the above advantages to quickly collect natural sweat from the hands. Tang et al. [116][61] designed a sensor that can rapidly detect cortisol through fingertip contact, using a highly permeable, porous PVA hydrogel. Its detection results can be obtained within 3.5 min of fingertip contact. Patches utilizing PVA hydrogel also exhibit excellent resistance to staining [106][96].
Sweat Transport and Management: When continuously monitoring sweat, it is important to consider how to prevent the accumulation of old sweat, as residual sweat can saturate the device and affect its long-term performance. Saha et al. [190][89] introduced an osmotic wearable for lactate sensing in sweat (OWLSS) that is capable of continuous monitoring, utilizing a specially designed paper channel to effectively manage sweat. Adding an evaporation pad to the channel and creating a serpentine channel path to increase the distance sweat must travel before reaching the evaporation pad can help delay channel saturation. Increasing the area of the evaporation pad also ensures that sweat evaporates faster than it flows into the channel under all conditions, allowing for continuous monitoring.

3.2. Signal Transmission and Power Management

Wearable sweat sensors require real-time monitoring over extended periods of time, necessitating the use of wireless signal transmission. Technologies such as Bluetooth, NFC, and RFID are commonly employed in the electronic systems of wearable sweat sensors due to their advantages of miniaturization and low power consumption. The communication and control of wearable sensors consume energy, particularly in the signal processing and wireless transmission circuits, such as those utilizing Bluetooth or radio frequency transmission [200,201,202][97][98][99]. However, the traditional battery power supply method hinders the continuous operation of the sensor on the skin due to its difficulty in deformation, poor battery life, and the presence of harmful substances. To address the issue of energy consumption, new battery materials suitable for use in wearable devices have been developed. In their study, Liu et al. [203][100] mentioned the use of super-resilient hard carbon nanofabrics (s-HCNFs) as carbonaceous anode materials. This high-capacity material exhibits excellent mechanical stability under twisting and folding, and its properties can be further enhanced by designing it as a 3D micro- or nanolayered structure. These properties make s-HCNFs well-suited for use in the fabrication of flexible wearable devices. Moreover, researchers are also applying scientific research results such as self-generation, wireless power supply, and flexible batteries to integrate them into wearable sensors.
NFC: The advantages of NFC include low power consumption, few components, and small size. Only small coils and NFC chips are required to achieve wireless signal transmission [204][101]. NFC chips and sensors can be magnetically coupled to form a stable system [90][83]. The modulated antenna can also provide wireless energy supply [35][102]. Cheng et al. [198][103] developed a sweat cortisol sensor that uses NFC for wireless communication and power. When detection is required, the NFC antenna is powered after the smartphone approaches the patch to power the circuit and begin measurement. The performance of the NFC antenna is also verified under different radii of curvature to ensure that the NFC chip can still provide stable power when the patch is bent.
Biofuels and Sweat-Activated Batteries: Sweat has the potential to provide electricity support for epidermal electronics as a sustainable bioenergy source [205,206][104][105]. Biofuel cells provide power based on the bioenergy in sweat. Yu et al. [200][97] report on a lactate biofuel cell based on full perspiration-powered electronic skin (PPES). The enzyme-catalyzed batteries exhibit a higher current density and stability. The electrolytes in sweat (Na+, K+, Cl) can also be used for electrochemical energy storage. Chen et al. [207][106] designed an array of sector electrodes (using copper and zinc thin films) to create a sweat-based energy generator based on redox reactions. Manjakkal et al. [208][107] report a supercapacitor using PEDOT: PSS as the active electrode and current collector. This material has been used in sensitive electrodes of electrochemical sensors [6][70] due to its high conductivity, large voltage window, and good mechanical properties.
In response to the need for flexibility, Liu et al. [199][108] developed a flexible sweat-activated battery that can be directly integrated onto the skin. Soft silicone is used for fixing and loading functional materials as a package housing. Two Nylon fabric bags containing electrolyte are used to stabilize the output voltage under various deformation conditions.
Triboelectric Nanogenerator: Since the invention of triboelectric nanogenerators (TENGs), many sensor platforms incorporating TENGs have been reported [209,210,211][109][110][111] due to their advantages of a high output voltage, power, low cost, and ease of fabrication [212,213,214,215][112][113][114][115]. In sweat-sensing applications, methods for optimizing TENGs are also being explored. Zahed et al. [216][116] used a PVDF/Co3O4 nanofiber-based TENG and a miniature Halbach magnet array for EMG to improve its electrical energy conversion efficiency. Baro et al. [217][117] proposed a textile-based sweat sensor incorporating a ZnO-based unipolar friction nanogenerator (STENG). Their study experimentally verified that hydrated salts in sweat promote an increase in conduction band electrons in ZnO, allowing the integrated STENG to output higher voltages. TENGs can also be integrated with flexible supercapacitors (SCs) to form a distributed power system, further enhancing the power-management capabilities of wearable devices [218][118]. The construction of self-charging power systems (SCPS) for TENG-SCs requires consideration of two key factors. Firstly, the AC power output by the TENG cannot be directly fed into the SC and must be converted using a rectifier. Secondly, it is difficult to share electrodes between the TENG and the SC, and alternative solutions such as shared substrates or packages are typically used [219][119]. These can include fabrics [220][120], thin films [221][121], carbon cloth [222[122][123],223], hydrogels [224][124], and others. The integration of TENGs with energy-storage technologies has led to the development of uninterrupted power supply (UPS) systems for wearable devices [225][125]. These systems provide a viable solution for the continuous power supply of wearable devices.
Solar Power: Singh et al. [226][126] installed a solar panel on top of a sweat sensor in the form of a smart watch to collect energy. A circular solar panel with a diameter of 2.2 inches was mounted on the top of the device as an energy source to power the integrated circuit of the smart watch. This combination provides a new idea for the power supply scheme of wearable devices.
Inertial Energy Harvesters: Inertial energy harvesters can convert the kinetic energy generated by human motion into electrical energy to power devices. These harvesters contain a proof mass that is excited by human motion, with the inertial energy of the motion being converted into electrical energy through piezoelectric, electrostatic, or electromagnetic methods [227,228][127][128]. To address the power density limitation imposed by the volume of the proof mass, Cai et al. [229][129] proposed an alternative approach. They utilized a planetary structure with a power-generating unit consisting of a base, coils, rotor, and magnets as an eccentric mass, thereby avoiding the use of a proof mass. In walking tests, the power output of this design reached 1.84 mW at the wrist and 2.95 mW at the ankle. The output of energy harvesters is influenced by factors such as the frequency of human movement and the placement of the device. To address this, Hoareau et al. [230][130] proposed a method for determining the optimal placement of cantilevered piezoelectric generators (PEGs). This method uses electromechanical modeling and accelerometer data to identify the optimal power source for cantilevered PEGs. According to the scenario presented in the article, the optimal placement for the device is along the axis normal to the surface of the right hand. Additionally, extremity segments such as the feet, hands, and forearms were identified as better energy sources. Beach et al. [231][131] compared the amount of energy harvested from the wrist, hip, ankle, and foot. Based on their analysis of the spectral content, optimal harvester parameters, and the average power output, it was concluded that the foot is able to provide more power, utilize a larger area, and exhibit a better broadband response during walking. Additionally, the foot was found to be least affected by changes in walking speed. To further improve energy harvesting, Sandhu et al. [232][132] proposed combining solar and kinetic energy harvesting as both energy and activity sensors.
Thermoelectricity: Thermoelectric systems convert the temperature difference (ΔT) between two interfaces into electricity. The ΔT between the human body and the environment is generally stable, making thermoelectric generators (TEGs) a promising power for providing a stable energy supply. However, TEGs require a high temperature difference [233][133] or a booster [234][134] to provide sufficient voltage, which unfortunately reduces the flexibility of portable devices and increases the complexity of the system. To solve the issues mentioned above, fiber-based TEGs can be structurally optimized to increase the output voltage [235,236][135][136]. The all-fiber thermoelectric sensing device proposed by Qing et al. [166][54] integrates the fiber-based organic electrochemical transistor (FOECT) and thermoelectric fiber (TEF). Such a power supply has sufficient output and does not require energy storage devices.
Despite promising research into self-powered modules, both biofuel cells and energy harvesters still face challenges in terms of energy efficiency, cell size, and durability. As a result, the dominance of lithium batteries remains difficult to challenge. A long-term vision and sustained efforts are required to fully transition from traditional rigid batteries to flexible power sources suitable for wearable systems.

References

  1. Parlak, O.; Keene, S.T.; Marais, A.; Curto, V.F.; Salleo, A. Molecularly selective nanoporous membrane-based wearable organic electrochemical device for noninvasive cortisol sensing. Sci. Adv. 2018, 4, eaar2904.
  2. Mei, X.; Yang, J.; Liu, J.; Li, Y. Wearable, nanofiber-based microfluidic systems with integrated electrochemical and colorimetric sensing arrays for multiplex sweat analysis. Chem. Eng. J. 2023, 454, 140248.
  3. Zhai, Q.; Yap, L.W.; Wang, R.; Gong, S.; Guo, Z.; Liu, Y.; Lyu, Q.; Wang, J.; Simon, G.P.; Cheng, W. Vertically Aligned Gold Nanowires as Stretchable and Wearable Epidermal Ion-Selective Electrode for Noninvasive Multiplexed Sweat Analysis. Anal. Chem. 2020, 92, 4647–4655.
  4. Lin, Y.-C.; Rinawati, M.; Chang, L.-Y.; Wang, Y.-X.; Wu, Y.-T.; Yen, Y.-H.; Chen, K.-J.; Ho, K.-C.; Yeh, M.-H. A non-invasive wearable sweat biosensor with a flexible N-GQDs/PANI nanocomposite layer for glucose monitoring. Sens. Actuators B 2023, 383, 133617.
  5. Sun, T.; Hui, J.; Zhou, L.; Lin, B.; Sun, H.; Bai, Y.; Zhao, J.; Mao, H. A low-cost and simple-fabricated epidermal sweat patch based on “cut-and-paste” manufacture. Sens. Actuators B 2022, 368, 132184.
  6. Tao, T.; Zhou, Y.; Ma, M.; He, H.; Gao, N.; Cai, Z.; Chang, G.; He, Y. Novel graphene electrochemical transistor with ZrO2/rGO nanocomposites functionalized gate electrode for ultrasensitive recognition of methyl parathion. Sens. Actuators B 2021, 328, 128936.
  7. Tao, T.; Gao, N.; He, H.; Zhou, R.; Tu, B.; Cai, Z.; Chang, G.; He, Y.; Ji, X. Au-PEDOT/rGO nanocomposites functionalized graphene electrochemical transistor for ultra-sensitive detection of acetaminophen in human urine. Anal. Chim. Acta 2022, 1191, 339306.
  8. Thanh, T.D.; Balamurugan, J.; Lee, S.H.; Kim, N.H.; Lee, J.H. Effective seed-assisted synthesis of gold nanoparticles anchored nitrogen-doped graphene for electrochemical detection of glucose and dopamine. Biosens. Bioelectron. 2016, 81, 259–267.
  9. Gao, N.; He, C.; Ma, M.; Cai, Z.; Zhou, Y.; Chang, G.; Wang, X.; He, Y. Electrochemical co-deposition synthesis of Au-ZrO2-graphene nanocomposite for a nonenzymatic methyl parathion sensor. Anal. Chim. Acta 2019, 1072, 25–34.
  10. Yu, Z.; Wu, H.; Xu, Z.; Yang, Z.; Lv, J.; Kong, C. Wearable Noninvasive Glucose Sensor Based on CuxO NFs/Cu NPs Nanocomposites. Sensors 2023, 23, 695.
  11. Asaduzzaman, M.; Zahed, M.A.; Sharifuzzaman, M.; Reza, M.S.; Hui, X.; Sharma, S.; Shin, Y.D.; Park, J.Y. A hybridized nano-porous carbon reinforced 3D graphene-based epidermal patch for precise sweat glucose and lactate analysis. Biosens. Bioelectron. 2022, 219, 114846.
  12. Gao, N.; Zhou, R.; Tu, B.; Tao, T.; Song, Y.; Cai, Z.; He, H.; Chang, G.; Wu, Y.; He, Y. Graphene electrochemical transistor incorporated with gel electrolyte for wearable and non-invasive glucose monitoring. Anal. Chim. Acta 2023, 1239, 340719.
  13. Lee, H.; Choi, T.K.; Lee, Y.B.; Cho, H.R.; Ghaffari, R.; Wang, L.; Choi, H.J.; Chung, T.D.; Lu, N.; Hyeon, T.; et al. A graphene-based electrochemical device with thermoresponsive microneedles for diabetes monitoring and therapy. Nat. Nanotechnol. 2016, 11, 566–572.
  14. Torrente-Rodriguez, R.M.; Tu, J.; Yang, Y.; Min, J.; Wang, M.; Song, Y.; Yu, Y.; Xu, C.; Ye, C.; IsHak, W.W.; et al. Investigation of cortisol dynamics in human sweat using a graphene-based wireless mHealth system. Matter 2020, 2, 921–937.
  15. Chen, Y.; Li, G.; Mu, W.; Wan, X.; Lu, D.; Gao, J.; Wen, D. Nonenzymatic Sweat Wearable Uric Acid Sensor Based on N-Doped Reduced Graphene Oxide/Au Dual Aerogels. Anal. Chem. 2023, 95, 3864–3872.
  16. Wang, Y.; Chen, F.; Ye, J.; Liu, H.; Zhang, T.; Li, Z. Reduced graphene oxide cotton fabric based on copper nanowires for flexible non-enzyme glucose sensor. Cellulose 2023, 30, 5131–5143.
  17. Wang, M.; Yang, Y.; Min, J.; Song, Y.; Tu, J.; Mukasa, D.; Ye, C.; Xu, C.; Heflin, N.; McCune, J.S.; et al. A wearable electrochemical biosensor for the monitoring of metabolites and nutrients. Nat. Biomed. Eng. 2022, 6, 1225–1235.
  18. Mukasa, D.; Wang, M.; Min, J.; Yang, Y.; Solomon, S.A.; Han, H.; Ye, C.; Gao, W. A Computationally Assisted Approach for Designing Wearable Biosensors toward Non-Invasive Personalized Molecular Analysis. Adv. Mater. 2023, 2212161.
  19. Tu, J.; Min, J.; Song, Y.; Xu, C.; Li, J.; Moore, J.; Hanson, J.; Hu, E.; Parimon, T.; Wang, T.Y.; et al. A wireless patch for the monitoring of C-reactive protein in sweat. Nat. Biomed. Eng. 2023, 14, 15.
  20. Huang, L.; Su, J.; Song, Y.; Ye, R. Laser-Induced Graphene: En Route to Smart Sensing. Nanomicro Lett 2020, 12, 157.
  21. Feng, J.; Jiang, Y.; Wang, K.; Li, J.; Zhang, J.; Tian, M.; Chen, G.; Hu, L.; Zhan, Y.; Qin, Y. An Energy-Efficient Flexible Multi-Modal Wireless Sweat Sensing System Based on Laser Induced Graphene. Sensors 2023, 23, 4818.
  22. Wang, Y.; Guo, H.; Yuan, M.; Yu, J.; Wang, Z.; Chen, X. One-step laser synthesis platinum nanostructured 3D porous graphene: A flexible dual-functional electrochemical biosensor for glucose and pH detection in human perspiration. Talanta 2023, 257, 124362.
  23. Naguib, M.; Kurtoglu, M.; Presser, V.; Lu, J.; Niu, J.; Heon, M.; Hultman, L.; Gogotsi, Y.; Barsoum, M.W. Two-dimensional nanocrystals produced by exfoliation of Ti3AlC2. Adv. Mater. 2011, 23, 4248–4253.
  24. VahidMohammadi, A.; Rosen, J.; Gogotsi, Y. The world of two-dimensional carbides and nitrides (MXenes). Science 2021, 372, eabf1581.
  25. Zhao, Q.-N.; Zhang, Y.-J.; Duan, Z.-H.; Wang, S.; Liu, C.; Jiang, Y.-D.; Tai, H.-L. A review on Ti3C2Tx-based nanomaterials: Synthesis and applications in gas and humidity sensors. Rare Met. 2020, 40, 1459–1476.
  26. Zhang, Y.; Wang, L.; Zhang, N.; Zhou, Z. Adsorptive environmental applications of MXene nanomaterials: A review. RSC Adv. 2018, 8, 19895–19905.
  27. Sharifuzzaman, M.; Zahed, M.A.; Reza, M.S.; Asaduzzaman, M.; Jeong, S.; Song, H.; Kim, D.K.; Zhang, S.; Park, J.Y. MXene/Fluoropolymer-Derived Laser-Carbonaceous All-Fibrous Nanohybrid Patch for Soft Wearable Bioelectronics. Adv. Funct. Mater. 2023, 33, 2208894.
  28. Magesh, V.; Sundramoorthy, A.K.; Ganapathy, D.; Atchudan, R.; Arya, S.; Alshgari, R.A.; Aljuwayid, A.M. Palladium Hydroxide (Pearlman’s Catalyst) Doped MXene (Ti(3)C(2)Tx) Composite Modified Electrode for Selective Detection of Nicotine in Human Sweat. Biosensors 2022, 13, 54.
  29. Li, Q.-F.; Chen, X.; Wang, H.; Liu, M.; Peng, H.-L. Pt/MXene-Based Flexible Wearable Non-Enzymatic Electrochemical Sensor for Continuous Glucose Detection in Sweat. ACS Appl. Mater. Interfaces 2023, 15, 13290–13298.
  30. Wang, L.; Xu, T.; He, X.; Zhang, X. Flexible, self-healable, adhesive and wearable hydrogel patch for colorimetric sweat detection. J. Mater. Chem. C 2021, 9, 14938–14945.
  31. Yang, C.; Suo, Z. Hydrogel ionotronics. Nat. Rev. Mater. 2018, 3, 125–142.
  32. Tringides, C.M.; Vachicouras, N.; de Lázaro, I.; Wang, H.; Trouillet, A.; Seo, B.R.; Elosegui-Artola, A.; Fallegger, F.; Shin, Y.; Casiraghi, C.; et al. Viscoelastic surface electrode arrays to interface with viscoelastic tissues. Nat. Nanotechnol. 2021, 16, 1019–1029.
  33. Daly, A.C.; Riley, L.; Segura, T.; Burdick, J.A. Hydrogel microparticles for biomedical applications. Nat. Rev. Mater. 2020, 5, 20–43.
  34. Xu, Z.; Zhou, F.; Yan, H.; Gao, G.; Li, H.; Li, R.; Chen, T. Anti-freezing organohydrogel triboelectric nanogenerator toward highly efficient Flex. human-machine interaction at −30 °C. Nano Energy 2021, 90, 106614.
  35. Duan, X.; Yu, J.; Zhu, Y.; Zheng, Z.; Liao, Q.; Xiao, Y.; Li, Y.; He, Z.; Zhao, Y.; Wang, H.; et al. Large-Scale Spinning Approach to Engineering Knittable Hydrogel Fiber for Soft Robots. ACS Nano 2020, 14, 14929–14938.
  36. Wang, L.; Zhou, M.; Xu, T.; Zhang, X. Multifunctional hydrogel as wound dressing for intelligent wound monitoring. Chem. Eng. J. 2022, 433, 134625.
  37. Liang, Y.; He, J.; Guo, B. Functional Hydrogels as Wound Dressing to Enhance Wound Healing. ACS Nano 2021, 15, 12687–12722.
  38. Wang, L.; Wang, J.; Fan, C.; Xu, T.; Zhang, X. Skin-like hydrogel-elastomer based electrochemical device for comfortable wearable biofluid monitoring. Chem. Eng. J. 2023, 455, 140609.
  39. Gao, N.; Cai, Z.; Chang, G.; He, Y. Non-invasive and wearable glucose biosensor based on gel electrolyte for detection of human sweat. J. Mater. Sci. 2023, 58, 890–901.
  40. Sarkar, R.; Farghaly, A.A.; Arachchige, I.U. Oxidative Self-Assembly of Au/Ag/Pt Alloy Nanoparticles into High-Surface Area, Mesoporous, and Conductive Aerogels for Methanol Electro-oxidation. Chem. Mater. 2022, 34, 5874–5887.
  41. Wang, C.; Gao, W.; Wan, X.; Yao, B.; Mu, W.; Gao, J.; Fu, Q.; Wen, D. In situ electrochemical synthesis of Pd aerogels as highly efficient anodic electrocatalysts for alkaline fuel cells. Chem. Sci. 2022, 13, 13956–13965.
  42. Du, R.; Joswig, J.-O.; Fan, X.; Hübner, R.; Spittel, D.; Hu, Y.; Eychmüller, A. Disturbance-Promoted Unconventional and Rapid Fabrication of Self-Healable Noble Metal Gels for (Photo-)Electrocatalysis. Matter 2020, 2, 908–920.
  43. Gao, W.; Wen, D. Recent advances of noble metal aerogels in biosensing. View 2021, 2, 20200124.
  44. Li, G.; Wang, C.; Chen, Y.; Liu, F.; Fan, H.; Yao, B.; Hao, J.; Yu, Y.; Wen, D. Dual Structural Design of Platinum-Nickel Hydrogels for Wearable Glucose Biosensing with Ultrahigh Stability. Small 2023, 19, e2206868.
  45. Keene, S.T.; Fogarty, D.; Cooke, R.; Casadevall, C.D.; Salleo, A.; Parlak, O. Wearable Organic Electrochemical Transistor Patch for Multiplexed Sensing of Calcium and Ammonium Ions from Human Perspiration. Adv. Healthcare Mater. 2019, 8, e1901321.
  46. Aerathupalathu Janardhanan, J.; Chen, Y.L.; Liu, C.T.; Tseng, H.S.; Wu, P.I.; She, J.W.; Hsiao, Y.S.; Yu, H.H. Sensitive Detection of Sweat Cortisol Using an Organic Electrochemical Transistor Featuring Nanostructured Poly(3,4-Ethylenedioxythiophene) Derivatives in the Channel Layer. Anal. Chem. 2022, 94, 7584–7593.
  47. Khodagholy, D.; Rivnay, J.; Sessolo, M.; Gurfinkel, M.; Leleux, P.; Jimison, L.H.; Stavrinidou, E.; Herve, T.; Sanaur, S.; Owens, R.M.; et al. High transconductance organic electrochemical transistors. Nat. Commun. 2013, 4, 2133.
  48. Wang, N.; Liu, Y.; Fu, Y.; Yan, F. AC Measurements Using Organic Electrochemical Transistors for Accurate Sensing. ACS Appl. Mater. Interfaces 2018, 10, 25834–25840.
  49. Li, Y.; Cui, B.; Zhang, S.; Li, B.; Li, J.; Liu, S.; Zhao, Q. Ion-Selective Organic Electrochemical Transistors: Recent Progress and Challenges. Small 2022, 18, e2107413.
  50. Sessolo, M.; Rivnay, J.; Bandiello, E.; Malliaras, G.G.; Bolink, H.J. Ion-selective organic electrochemical transistors. Adv. Mater. 2014, 26, 4803–4807.
  51. Coppedè, N.; Giannetto, M.; Villani, M.; Lucchini, V.; Battista, E.; Careri, M.; Zappettini, A. Ion selective textile organic electrochemical transistor for wearable sweat monitoring. Org. Electron. 2020, 78, 105579.
  52. Tao, Y.; Zhu, R.; Hao, P.; Jiang, W.; Li, M.; Liu, Q.; Yang, L.; Wang, Y.; Wang, D. Textile-based dual-mode organic electrochemical transistors for lactate biosensing. Mater. Sci. Eng. B 2023, 290, 116356.
  53. Fang, Y.; Feng, J.; Shi, X.; Yang, Y.; Wang, J.; Sun, X.; Li, W.; Sun, X.; Peng, H. Coaxial fiber organic electrochemical transistor with high transconductance. Nano Res. 2023.
  54. Qing, X.; Chen, H.; Zeng, F.; Jia, K.; Shu, Q.; Wu, J.; Xu, H.; Lei, W.; Liu, D.; Wang, X.; et al. All-Fiber Integrated Thermoelectrically Powered Physiological Monitoring Biosensor. Adv. Fiber Mater. 2023, 5, 1025–1036.
  55. Qu, Y.; Nguyen-Dang, T.; Page, A.G.; Yan, W.; Das Gupta, T.; Rotaru, G.M.; Rossi, R.M.; Favrod, V.D.; Bartolomei, N.; Sorin, F. Superelastic Multimaterial Electronic and Photonic Fibers and Devices via Thermal Drawing. Adv. Mater. 2018, 30, 1707251.
  56. Zhang, T.; Wang, Z.; Srinivasan, B.; Wang, Z.; Zhang, J.; Li, K.; Boussard-Pledel, C.; Troles, J.; Bureau, B.; Wei, L. Ultraflexible Glassy Semiconductor Fibers for Thermal Sensing and Positioning. ACS Appl. Mater. Interfaces 2019, 11, 2441–2447.
  57. Wu, J.; Sato, Y.; Guo, Y. Microelectronic fibers for multiplexed sweat sensing. Anal. Bioanal. Chem. 2023, 415, 4307–4318.
  58. Garcia-Rey, S.; Gil-Hernandez, E.; Basabe-Desmonts, L.; Benito-Lopez, F. Colorimetric Determination of Glucose in Sweat Using an Alginate-Based Biosystem. Polymers 2023, 15, 1218.
  59. Garcia-Rey, S.; Ojeda, E.; Gunatilake, U.B.; Basabe-Desmonts, L.; Benito-Lopez, F. Alginate Bead Biosystem for the Determination of Lactate in Sweat Using Image Analysis. Biosensors 2021, 11, 379.
  60. Gunatilake, U.B.; Garcia-Rey, S.; Ojeda, E.; Basabe-Desmonts, L.; Benito-Lopez, F. TiO2 Nanotubes Alginate Hydrogel Scaffold for Rapid Sensing of Sweat Biomarkers: Lactate and Glucose. ACS Appl. Mater. Interfaces 2021, 13, 37734–37745.
  61. Tang, W.; Yin, L.; Sempionatto, J.R.; Moon, J.M.; Teymourian, H.; Wang, J. Touch-Based Stressless Cortisol Sensing. Adv. Mater. 2021, 33, e2008465.
  62. Tang, Y.; Gan, S.; Zhong, L.; Sun, Z.; Xu, L.; Liao, C.; Lin, K.; Cui, X.; He, D.; Ma, Y.; et al. Lattice Proton Intercalation to Regulate WO3-Based Solid-Contact Wearable pH Sensor for Sweat Analysis. Adv. Funct. Mater. 2021, 32, 2107653.
  63. Windmiller, J.R.; Bandodkar, A.J.; Valdes-Ramirez, G.; Parkhomovsky, S.; Martinez, A.G.; Wang, J. Electrochemical sensing based on printable temporary transfer tattoos. Chem. Commun. 2012, 48, 6794–6796.
  64. Guinovart, T.; Bandodkar, A.J.; Windmiller, J.R.; Andrade, F.J.; Wang, J. A potentiometric tattoo sensor for monitoring ammonium in sweat. Analyst 2013, 138, 7031–7038.
  65. Jia, W.; Bandodkar, A.J.; Valdés-Ramírez, G.; Windmiller, J.R.; Yang, Z.; Ramírez, J.; Chan, G.; Wang, J. Electrochemical Tattoo Biosensors for Real-Time Noninvasive Lactate Monitoring in Human Perspiration. Anal. Chem. 2013, 85, 6553–6560.
  66. Bandodkar, A.J.; Molinnus, D.; Mirza, O.; Guinovart, T.; Windmiller, J.R.; Valdes-Ramirez, G.; Andrade, F.J.; Schoning, M.J.; Wang, J. Epidermal tattoo potentiometric sodium sensors with wireless signal transduction for continuous non-invasive sweat monitoring. Biosens. Bioelectron. 2014, 54, 603–609.
  67. Katseli, V.; Economou, A.; Kokkinos, C. Smartphone-Addressable 3D-Printed Electrochemical Ring for Nonenzymatic Self-Monitoring of Glucose in Human Sweat. Anal. Chem. 2021, 93, 3331–3336.
  68. Anastasova, S.; Crewther, B.; Bembnowicz, P.; Curto, V.; Ip, H.M.; Rosa, B.; Yang, G.Z. A wearable multisensing patch for continuous sweat monitoring. Biosens. Bioelectron. 2017, 93, 139–145.
  69. Nyein, H.Y.Y.; Tai, L.C.; Ngo, Q.P.; Chao, M.; Zhang, G.B.; Gao, W.; Bariya, M.; Bullock, J.; Kim, H.; Fahad, H.M.; et al. A Wearable Microfluidic Sensing Patch for Dynamic Sweat Secretion Analysis. ACS Sens. 2018, 3, 944–952.
  70. Gao, W.; Emaminejad, S.; Nyein, H.Y.Y.; Challa, S.; Chen, K.; Peck, A.; Fahad, H.M.; Ota, H.; Shiraki, H.; Kiriya, D.; et al. Fully integrated wearable sensor arrays for multiplexed in situ perspiration analysis. Nature 2016, 529, 509–514.
  71. Nyein, H.Y.; Gao, W.; Shahpar, Z.; Emaminejad, S.; Challa, S.; Chen, K.; Fahad, H.M.; Tai, L.C.; Ota, H.; Davis, R.W.; et al. A Wearable Electrochemical Platform for Noninvasive Simultaneous Monitoring of Ca(2+) and pH. ACS Nano 2016, 10, 7216–7224.
  72. Sempionatto, J.R.; Lin, M.; Yin, L.; De la Paz, E.; Pei, K.; Sonsa-Ard, T.; de Loyola Silva, A.N.; Khorshed, A.A.; Zhang, F.; Tostado, N.; et al. An epidermal patch for the simultaneous monitoring of haemodynamic and metabolic biomarkers. Nat. Biomed. Eng. 2021, 5, 737–748.
  73. Gao, W.; Nyein, H.Y.Y.; Shahpar, Z.; Fahad, H.M.; Chen, K.; Emaminejad, S.; Gao, Y.; Tai, L.-C.; Ota, H.; Wu, E.; et al. Wearable Microsensor Array for Multiplexed Heavy Metal Monitoring of Body Fluids. ACS Sens. 2016, 1, 866–874.
  74. Emaminejad, S.; Gao, W.; Wu, E.; Davies, Z.A.; Yin Yin Nyein, H.; Challa, S.; Ryan, S.P.; Fahad, H.M.; Chen, K.; Shahpar, Z.; et al. Autonomous sweat extraction and analysis applied to cystic fibrosis and glucose monitoring using a fully integrated wearable platform. Proc. Natl. Acad. Sci. USA 2017, 114, 4625–4630.
  75. Kim, J.; Gutruf, P.; Chiarelli, A.M.; Heo, S.Y.; Cho, K.; Xie, Z.; Banks, A.; Han, S.; Jang, K.-I.; Lee, J.W.; et al. Miniaturized Battery-Free Wireless Systems for Wearable Pulse Oximetry. Adv. Funct. Mater. 2017, 27, 1604373.
  76. Chen, H.; Bao, S.; Lu, C.; Wang, L.; Ma, J.; Wang, P.; Lu, H.; Shu, F.; Oetomo, S.B.; Chen, W. Design of an Integrated Wearable Multi-Sensor Platform Based on Flexible Materials for Neonatal Monitoring. IEEE Access 2020, 8, 23732–23747.
  77. Wang, J.; Wang, L.; Li, G.; Yan, D.; Liu, C.; Xu, T.; Zhang, X. Ultra-Small Wearable Flexible Biosensor for Continuous Sweat Analysis. ACS Sens. 2022, 7, 3102–3107.
  78. He, X.; Xu, T.; Gu, Z.; Gao, W.; Xu, L.-P.; Pan, T.; Zhang, X. Flexible and Superwettable Bands as a Platform toward Sweat Sampling and Sensing. Anal. Chem. 2019, 91, 4296–4300.
  79. Steijlen, A.S.M.; Jansen, K.M.B.; Bastemeijer, J.; French, P.J.; Bossche, A. Low-Cost Wearable Fluidic Sweat Collection Patch for Continuous Analyte Monitoring and Offline Analysis. Anal. Chem. 2022, 94, 6893–6901.
  80. Koh, A.; Kang, D.; Xue, Y.; Lee, S.; Pielak, R.M.; Kim, J.; Hwang, T.; Min, S.; Banks, A.; Bastien, P.; et al. A soft, wearable microfluidic device for the capture, storage, and colorimetric sensing of sweat. Sci. Transl. Med. 2016, 8, ra165–ra366.
  81. Martin, A.; Kim, J.; Kurniawan, J.F.; Sempionatto, J.R.; Moreto, J.R.; Tang, G.; Campbell, A.S.; Shin, A.; Lee, M.Y.; Liu, X.; et al. Epidermal Microfluidic Electrochemical Detection System: Enhanced Sweat Sampling and Metabolite Detection. ACS Sens. 2017, 2, 1860–1868.
  82. Nyein, H.Y.Y.; Bariya, M.; Kivimäki, L.; Uusitalo, S.; Liaw, T.S.; Jansson, E.; Ahn, C.H.; Hangasky, J.A.; Zhao, J.; Lin, Y.; et al. Regional and correlative sweat analysis using high-throughput microfluidic sensing patches toward decoding sweat. Sci. Adv. 2019, 5, eaaw9906.
  83. Bandodkar, A.J.; Gutruf, P.; Choi, J.; Lee, K.; Sekine, Y.; Reeder, J.T.; Jeang, W.J.; Aranyosi, A.J.; Lee, S.P.; Model, J.B.; et al. Battery-free, skin-interfaced microfluidic/electronic systems for simultaneous electrochemical, colorimetric, and volumetric analysis of sweat. Sci. Adv. 2019, 5, eaav3294.
  84. Huang, X.; Li, J.; Liu, Y.; Wong, T.; Su, J.; Yao, K.; Zhou, J.; Huang, Y.; Li, H.; Li, D.; et al. Epidermal self-powered sweat sensors for glucose and lactate monitoring. Bio-Des. Manuf. 2021, 5, 201–209.
  85. Kwon, K.; Kim, J.U.; Deng, Y.; Krishnan, S.R.; Choi, J.; Jang, H.; Lee, K.; Su, C.-J.; Yoo, I.; Wu, Y.; et al. An on-skin platform for wireless monitoring of flow rate, cumulative loss and temperature of sweat in real time. Nat. Electron. 2021, 4, 302–312.
  86. Paul Kunnel, B.; Demuru, S. An epidermal wearable microfluidic patch for simultaneous sampling, storage, and analysis of biofluids with counterion monitoring. Lab Chip 2022, 22, 1793–1804.
  87. Shi, H.H.; Cao, Y.; Zeng, Y.N.; Zhou, Y.N.; Wen, W.H.; Zhang, C.X.; Zhao, Y.L.; Chen, Z. Wearable tesla valve-based sweat collection device for sweat colorimetric analysis. Talanta 2022, 240, 10.
  88. Mei, X.; Yang, J.; Yu, X.; Peng, Z.; Zhang, G.; Li, Y. Wearable molecularly imprinted electrochemical sensor with integrated nanofiber-based microfluidic chip for in situ monitoring of cortisol in sweat. Sens. Actuators B 2023, 381, 133451.
  89. Saha, T.; Songkakul, T.; Knisely, C.T.; Yokus, M.A.; Daniele, M.A.; Dickey, M.D.; Bozkurt, A.; Velev, O.D. Wireless Wearable Electrochemical Sensing Platform with Zero-Power Osmotic Sweat Extraction for Continuous Lactate Monitoring. ACS Sens. 2022, 7, 2037–2048.
  90. Yang, M.; Sun, N.; Lai, X.; Wu, J.; Wu, L.; Zhao, X.; Feng, L. Paper-Based Sandwich-Structured Wearable Sensor with Sebum Filtering for Continuous Detection of Sweat pH. ACS Sens. 2023, 8, 176–186.
  91. Liang, B.; Cao, Q.; Mao, X.; Pan, W.; Tu, T.; Fang, L.; Ye, X. An Integrated Paper-Based Microfluidic Device for Real-Time Sweat Potassium Monitoring. IEEE Sens. J. 2021, 21, 9642–9648.
  92. Bolat, G.; De la Paz, E.; Azeredo, N.F.; Kartolo, M.; Kim, J.; de Loyola, E.S.A.N.; Rueda, R.; Brown, C.; Angnes, L.; Wang, J.; et al. Wearable soft electrochemical microfluidic device integrated with iontophoresis for sweat biosensing. Anal. Bioanal. Chem. 2022, 414, 5411–5421.
  93. Kim, J.; Jeerapan, I.; Imani, S.; Cho, T.N.; Bandodkar, A.; Cinti, S.; Mercier, P.P.; Wang, J. Noninvasive Alcohol Monitoring Using a Wearable Tattoo-Based Iontophoretic-Biosensing System. ACS Sens. 2016, 1, 1011–1019.
  94. Zhang, K.; Zhang, J.; Wang, F.; Kong, D. Stretchable and Superwettable Colorimetric Sensing Patch for Epidermal Collection and Analysis of Sweat. ACS Sens. 2021, 6, 2261–2269.
  95. Lin, P.H.; Sheu, S.C.; Chen, C.W.; Huang, S.C.; Li, B.R. Wearable hydrogel patch with noninvasive, electrochemical glucose sensor for natural sweat detection. Talanta 2022, 241, 11.
  96. Zhang, S.; Liu, Y.; Wang, J.; Liu, Z. A Laser-Induced Photoelectrochemical Sensor for Natural Sweat Cu2+ Detection. Chemosensors 2022, 10, 169.
  97. Yu, Y.; Nassar, J.; Xu, C.; Min, J.; Yang, Y.; Dai, A.; Doshi, R.; Huang, A.; Song, Y.; Gehlhar, R.; et al. Biofuel-powered soft electronic skin with multiplexed and wireless sensing for human-machine interfaces. Sci. Robot. 2020, 5, eaaz7946.
  98. Chen, J.; Huang, Y.; Zhang, N.; Zou, H.; Liu, R.; Tao, C.; Fan, X.; Wang, Z.L. Micro-cable structured textile for simultaneously harvesting solar and mechanical energy. Nat. Energy 2016, 1, 16138.
  99. Suarez, F.; Nozariasbmarz, A.; Vashaee, D.; Öztürk, M.C. Designing thermoelectric generators for self-powered wearable electronics. Energy Environ. Sci. 2016, 9, 2099–2113.
  100. Liu, S.; Kang, L.; Zhang, J.; Jun, S.C.; Yamauchi, Y. Carbonaceous Anode Materials for Non-aqueous Sodium- and Potassium-Ion Hybrid Capacitors. ACS Energy Lett. 2021, 6, 4127–4154.
  101. Anabtawi, N.; Freeman, S.; Ferzli, R. A fully implantable, NFC enabled, continuous interstitial glucose monitor. In Proceedings of the 2016 IEEE-EMBS International Conference on Biomedical and Health Informatics (BHI), Las Vegas, NV, USA, 24–27 February 2016; pp. 612–615.
  102. Rose, D.P.; Ratterman, M.E.; Griffin, D.K.; Hou, L.; Kelley-Loughnane, N.; Naik, R.R.; Hagen, J.A.; Papautsky, I.; Heikenfeld, J.C. Adhesive RFID Sensor Patch for Monitoring of Sweat Electrolytes. IEEE Trans. Biomed. Eng. 2015, 62, 1457–1465.
  103. Cheng, C.; Li, X.; Xu, G.; Lu, Y.; Low, S.S.; Liu, G.; Zhu, L.; Li, C.; Liu, Q. Battery-free, wireless, and flexible electrochemical patch for in situ analysis of sweat cortisol via near field communication. Biosens. Bioelectron. 2021, 172, 112782.
  104. Escalona-Villalpando, R.A.; Ortiz-Ortega, E.; Bocanegra-Ugalde, J.P.; Minteer, S.D.; Ledesma-Garcia, J.; Arriaga, L.G. Clean energy from human sweat using an enzymatic patch. J. Power Sources 2019, 412, 496–504.
  105. Bandodkar, A.J.; You, J.M.; Kim, N.H.; Gu, Y.; Kumar, R.; Mohan, A.M.V.; Kurniawan, J.; Imani, S.; Nakagawa, T.; Parish, B.; et al. Soft, stretchable, high power density electronic skin-based biofuel cells for scavenging energy from human sweat. Energy Environ. Sci. 2017, 10, 1581–1589.
  106. Chen, Y.; Xue, Y.; Liu, W.; Li, S.; Wang, X.; Zhou, W.; Zhang, G.; Liu, K.; Zhang, H.; Zhao, Y.; et al. Untethered artificial muscles powered by wearable sweat-based energy generator. Nano Today 2023, 49, 101765.
  107. Manjakkal, L.; Pullanchiyodan, A.; Yogeswaran, N.; Hosseini, E.S.; Dahiya, R. A Wearable Supercapacitor Based on Conductive PEDOT:PSS-Coated Cloth and a Sweat Electrolyte. Adv. Mater. 2020, 32, e1907254.
  108. Liu, Y.; Huang, X.; Zhou, J.; Yiu, C.K.; Song, Z.; Huang, W.; Nejad, S.K.; Li, H.; Wong, T.H.; Yao, K.; et al. Stretchable Sweat-Activated Battery in Skin-Integrated Electronics for Continuous Wireless Sweat Monitoring. Adv. Sci. 2022, 9, e2104635.
  109. Qin, Y.; Mo, J.; Liu, Y.; Zhang, S.; Wang, J.; Fu, Q.; Wang, S.; Nie, S. Stretchable Triboelectric Self-Powered Sweat Sensor Fabricated from Self-Healing Nanocellulose Hydrogels. Adv. Funct. Mater. 2022, 32, 2201846.
  110. Song, Y.; Min, J.H.; Yu, Y.; Wang, H.B.; Yang, Y.R.; Zhang, H.X.; Gao, W. Wireless battery-free wearable sweat sensor powered by human motion. Sci. Adv. 2020, 6, 10.
  111. Gai, Y.; Wang, E.; Liu, M.; Xie, L.; Bai, Y.; Yang, Y.; Xue, J.; Qu, X.; Xi, Y.; Li, L.; et al. A Self-Powered Wearable Sensor for Continuous Wireless Sweat Monitoring. Small Methods 2022, 6, 2200653.
  112. Lin, Z.; Chen, J.; Yang, J. Recent Progress in Triboelectric Nanogenerators as a Renewable and Sustainable Power Source. J. Nanomater. 2016, 2016, 5651613.
  113. Hu, Y.; Wang, Z.L. Recent progress in piezoelectric nanogenerators as a sustainable power source in self-powered systems and active sensors. Nano Energy 2015, 14, 3–14.
  114. Briscoe, J.; Dunn, S. Piezoelectric nanogenerators—A review of nanostructured piezoelectric energy harvesters. Nano Energy 2015, 14, 15–29.
  115. Hinchet, R.; Seung, W.; Kim, S.-W. Recent Progress on Flexible Triboelectric Nanogenerators for SelfPowered Electronics. ChemSusChem 2015, 8, 2327–2344.
  116. Zahed, M.A.; Rana, S.M.S.; Sharifuzzaman, M.; Jeong, S.; Pradhan, G.B.; Song, H.S.; Park, J.Y. A Hybrid Nanogenerator-Driven Self-Powered Wearable Perspiration Monitoring System. In Proceedings of the 2023 IEEE 36th International Conference on Micro Electro Mechanical Systems (MEMS), Munich, Germany, 15–19 January 2023; pp. 732–735.
  117. Baro, B.; Khimhun, S.; Das, U.; Bayan, S. ZnO based triboelectric nanogenerator on textile platform for wearable sweat sensing application. Nano Energy 2023, 108, 108212.
  118. Lu, Y.; Wu, T.; Ma, Z.; Mi, Y.; Zhao, Z.; Liu, F.; Cao, X.; Wang, N. Integration of Flexible Supercapacitors with Triboelectric Nanogenerators: A Review. Batteries 2023, 9, 281.
  119. Pu, X.; Hu, W.; Wang, Z.L. Toward Wearable Self-Charging Power Systems: The Integration of Energy-Harvesting and Storage Devices. Small 2018, 14, 1702817.
  120. Zhao, J.; Cong, Z.; Hu, J.; Lu, H.; Wang, L.; Wang, H.; Malyi, O.I.; Pu, X.; Zhang, Y.; Shao, H.; et al. Regulating zinc electroplating chemistry to achieve high energy coaxial fiber Zn ion supercapacitor for self-powered textile-based monitoring system. Nano Energy 2022, 93, 106893.
  121. Wang, L.; Wu, H.; Zhai, X.; Shi, J.; Zhou, Q.; Li, H.; Wan, J. Ti3C2Tx MXene/dopamine-modified polypyrrole flexible composite electrodes with application in energy storage devices. J. Alloys Compd. 2023, 946, 169347.
  122. Huang, Y.; Wang, L.; Li, X.; Yang, X.; Lü, W. Washable All-in-One Self-Charging Power Unit Based on a Triboelectric Nanogenerator and Supercapacitor for Smart Textiles. Langmuir 2023, 39, 8855–8864.
  123. Zhu, Z.; Liang, X.; Luo, H.; Wang, L.; Gao, Y.; Li, X.; Yang, X.; Lü, W. Flexible self-powered energy systems based on H2O/Ni2+ intercalated NixV2O5·nH2O. Chem. Eur. J. 2023, e202301583.
  124. Wang, Z.; Yao, S.; Wang, S.; Liu, Z.; Wan, X.; Hu, Q.; Zhao, Y.; Xiong, C.; Li, L. Self-powered energy harvesting and implantable storage system based on hydrogel-enabled all-solid-state supercapacitor and triboelectric nanogenerator. Chem. Eng. J. 2023, 463, 142427.
  125. Mi, Y.; Lu, Y.; Wang, X.; Zhao, Z.; Cao, X.; Wang, N. From Triboelectric Nanogenerator to Uninterrupted Power Supply System: The Key Role of Electrochemical Batteries and Supercapacitors. Batteries 2022, 8, 215.
  126. Singh, J.; Ning, B.; Lee, P.; Liu, L. A Solar-Driven Wearable Multiplexed Bio-Sensing System For Noninvasive Healthcare Monitoring In Sweat. In Proceedings of the 2023 IEEE 36th International Conference on Micro Electro Mechanical Systems (MEMS), Munich, Germany, 15–19 January 2023; pp. 440–443.
  127. Roundy, S.; Rantz, R.; Xue, T.; Halim, M.A. Inertial Energy Harvesting for Wearables. In Proceedings of the 2018 IEEE SENSORS, New Delhi, India, 28–31 October 2018; pp. 1–4.
  128. Mitcheson, P.D.; Yeatman, E.M.; Rao, G.K.; Holmes, A.S.; Green, T.C. Energy Harvesting From Human and Machine Motion for Wireless Electronic Devices. Proc. IEEE 2008, 96, 1457–1486.
  129. Cai, M.; Liao, W.H. High-Power Density Inertial Energy Harvester Without Additional Proof Mass for Wearables. IEEE Internet Things J. 2021, 8, 297–308.
  130. Hoareau, D.; Jodin, G.; Laaraibi, A.-r.A.; Prioux, J.; Razan, F. Available Kinetic Energy Sources on the Human Body during Sports Activities: A Numerical Approach Based on Accelerometers for Cantilevered Piezoelectric Harvesters. Energies 2023, 16, 2695.
  131. Beach, C.; Casson, A.J. Inertial Kinetic Energy Harvesters for Wearables: The Benefits of Energy Harvesting at the Foot. IEEE Access 2020, 8, 208136–208148.
  132. Sandhu, M.M.; Khalifa, S.; Geissdoerfer, K.; Jurdak, R.; Portmann, M.; Kusy, B. FusedAR: Energy-Positive Human Activity Recognition Using Kinetic and Solar Signal Fusion. IEEE Sens. J. 2023, 23, 12411–12426.
  133. Yang, Y.; Lin, Z.-H.; Hou, T.; Zhang, F.; Wang, Z.L. Nanowire-composite based flexible thermoelectric nanogenerators and self-powered temperature sensors. Nano Res. 2012, 5, 888–895.
  134. Kim, J.; Khan, S.; Wu, P.; Park, S.; Park, H.; Yu, C.; Kim, W. Self-charging wearables for continuous health monitoring. Nano Energy 2021, 79, 105419.
  135. Sun, T.; Zhou, B.; Zheng, Q.; Wang, L.; Jiang, W.; Snyder, G.J. Stretchable fabric generates electric power from woven thermoelectric fibers. Nat. Commun. 2020, 11, 572.
  136. Lee, J.A.; Aliev, A.E.; Bykova, J.S.; de Andrade, M.J.; Kim, D.; Sim, H.J.; Lepró, X.; Zakhidov, A.A.; Lee, J.-B.; Spinks, G.M.; et al. Woven-Yarn Thermoelectric Textiles. Adv. Mater. 2016, 28, 5038–5044.
More
Video Production Service