Next Article in Journal
A Ratiometric Selective Fluorescent Probe Derived from Pyrene for Cu2+ Detection
Next Article in Special Issue
Recent Advances of Fluorescence Probes for Imaging of Ferroptosis Process
Previous Article in Journal
Investigating Organic Vapor Sensing Properties of Composite Carbon Nanotube-Zinc Oxide Nanowire
Previous Article in Special Issue
Nanoparticle-Mediated Signaling for Aptamer-Based Multiplexed Detection of Cortisol and Neuropeptide Y in Serum
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Progresses in NIR-II Luminescent Bio/Chemo Sensors Based on Lanthanide Nanocrystals

1
College of Life and Environmental Science, Minzu University of China, Beijing 100081, China
2
College of Science, Minzu University of China, Beijing 100081, China
3
Key Laboratory of Luminescence and Optical Information, Ministry of Education, Institute of Optoelectronic Technology, Beijing Jiaotong University, Beijing 100044, China
4
School of Medical Imaging, Wannan Medical College, Wuhu 241002, China
*
Author to whom correspondence should be addressed.
Chemosensors 2022, 10(6), 206; https://doi.org/10.3390/chemosensors10060206
Submission received: 25 April 2022 / Revised: 20 May 2022 / Accepted: 23 May 2022 / Published: 30 May 2022

Abstract

:
Fluorescent bio/chemosensors are widely used in the field of biological research and medical diagnosis, with the advantages of non-invasiveness, high sensitivity, and good selectivity. In particular, luminescent bio/chemosensors, based on lanthanide nanocrystals (LnNCs) with a second near-infrared (NIR-II) emission, have attracted much attention, owing to greater penetration depth, aside from the merits of narrow emission band, abundant emission lines, and long lifetimes. In this review, NIR-II LnNCs-based bio/chemo sensors are summarized from the perspectives of the mechanisms of NIR-II luminescence, synthesis method of LnNCs, strategy of luminescence enhancement, sensing mechanism, and targeted bio/chemo category. Finally, the problems that exist in present LnNCs-based bio/chemosensors are discussed, and the future development trend is prospected.

Graphical Abstract

1. Introduction

Bio/chemical detection is of great significance in biological research and medical diagnosis [1,2]. So far, various detection methods have been developed, such as colorimetry [3], chromatography [4], electrochemical method [5], and fluorescence spectroscopy [6]. These methods have the disadvantages of complicated operation processes, low sensitivity, inability to perform real-time detection, and in vivo application compared with fluorescent spectroscopy. Fluorescent spectroscopy is a new type detection method, which has the advantages of high sensitivity, good selectivity, low detection limit, and high resolution [7,8]. Meanwhile, it may be applied to real-time monitoring of biological substances [9].
Fluorescent spectroscopy is divided into visible, NIR-I (700~900 nm), and NIR-II (1000~1700 nm), according to the different emission wavelengths [10,11,12,13]. Nevertheless, the autofluorescence generated (Figure 1a) [14], interference of scattering (Figure 1b) [14], and absorption (Figure 1c) [15] in the biological tissues severely limits the application of visible light and NIR-I in the biological body when the excitation and emission light pass through biological tissues [14,15]. In this wavelength range, especially, the scattering and autofluorescence of organisms are significant, forming a strong signal background and interference. To circumvent these restrictions, NIR-II has been emerging in recent years since it can dramatically reduce scattering lights (Figure 1d) [16] and increase penetration depth (Figure 1e) [17,18,19] in biological applications, compared with those emitting in the visible or the NIR-I. Therefore, the research on NIR-II luminescent materials has gradually increased in bioimaging and bio/chemical sensing due to the unique superiority of NIR-II luminescent materials (Figure 2).
The current study of NIR-II luminescent materials mainly includes the following: quantum dots (QDs) [20,21], single carbon nanotubes (SWCNTs) [22], organic fluorescent probe [23,24,25,26,27,28,29], and lanthanide nanocrystals (LnNCs) [30,31,32,33,34,35,36,37]. Most QDs contain heavy metal elements such as lead, mercury, or arsenic [20]. This potential biological toxicity severely limits the application of QDs in organisms. The low luminescence quantum yield (<0.1%) and poor biocompatibility of SWCNTs limit their application in NIR-II [22]. Organic probes generally face extremely low fluorescence quantum yield, poor water solubility, and are prone to photobleaching and fluorescence quenching, which greatly limit the application of organic fluorescent probes in vivo [23,24,25,26,27,28,29]. Meanwhile, the emission wavelength of organic probes is short, most of which are around 1000 nm, which severely limits its application in the second near-infrared region [27,29]. In contrast, lanthanide ions (Ln3+) have a unique 4f electronic structure [38,39,40]. The spectrum produced by the f-f transition has narrow-band emission characteristics and is not sensitive to the environment, and the emission only involves atomic transitions, so they have strong resistance to photobleaching [12,38,39,40]. Meanwhile, the Ln3+ can effectively protect the internal 4f orbitals from the interference of the external crystal field due to the 5s25sp6 electron orbitals, make Ln3+ have a longer luminescence lifetime, and effectively avoid the interference of the autofluorescence of biological tissues [41,42]. Therefore, LnNCs, as a new generation of NIR-II probes, with low photobleaching, long luminescence lifetimes, low long-term cytotoxicity, narrow emission band widths, chemical stability, and large Stokes shifts, have gained more attention recently for bioimaging applications and bio/chemo sensors.
At present, the reviews on LnNCs mainly focus on discussing the application of LnNCs for in vivo imaging [12,38,39,40,41,42], and the discussion of LnNCs in bio/chemo sensing is only briefly mentioned as part of the imaging application. Therefore, a comprehensive and detailed review of NIR-II LnNCs in bio/chemo sensing is of great significance for researchers to thoroughly understand the research progress of LnNCs in bio/chemo sensing, and to provide ideas for the design of NIR-II LnNCs sensing probes. In this review, the latest research progress on NIR-II LnNCs bio/chemo sensors has been retrospected. Firstly, the design and synthesis of NIR-II LnNCs are discussed from a spectral point of view. Secondly, various application fields and sensing mechanisms of bio/chemo sensors are categorized and commented. Finally, the disadvantages of NIR-II LnNCs chemo/bio sensors and their prospects are given.

2. Design and Synthesis of NIR-II LnNCs

2.1. Mechanisms of NIR-II Luminescence

In LnNCs, three components are commonly included: a host matrix, a sensitizer, and an activator [36,37,38,39,40,41,42]. The host material should meet the requirements of optical transparency and low lattice phonon energy. At present, fluoride is mainly used as the host. The sensitizers and activators are generally Ln3+. Nd3+, Er3+, and Yb3+ are demonstrated to be efficient sensitizers due to their large absorption cross-sections in NIR-I or NIR-II regions (Figure 3a) [36]. The absorption peaks of Nd3+ are located at 730 nm and 808 nm; the absorption peaks of Er3+ are located at 808 nm, 980 nm, and 1550 nm; the absorption peak of Yb3+ is located at 980 nm.
The activators with NIR-II emission mainly include Pr3+, Nd3+, Ho3+, Er3+, and Tm3+ (Figure 3a,b) [36,41]. To overcome the weak light absorption problem of the activator ion itself, sensitizers with higher absorption coefficients are co-doped into the host, serving to harvest the excitation photons and transfer the excitation energy to activators, thus populating the radiative transition of activators for NIR-II luminescence. Therefore, NIR-II luminescence is the mechanism of LnNCs: firstly, the excited states of the sensitizers and activators split into different energy levels, in a ladder-like manner, in the crystal field maintained by the host matrix. The ground state electrons of the sensitizer are excited to the excited state under the excitation light of the appropriate wavelength. Then, the energy is transferred to the activator, and the activator is excited to the excited state. Finally, the excited state electrons return to the ground state and emit NIR-II luminescence (Figure 3b). According to NIR-II emission of different activators, it can be roughly divided into three types of NIR-II probes [36,41,43,44]: (1) Er3+ based NIR-II probes. NIR-II emission at intense 1525~1550 nm can be generated through 4I13/24I15/2 radiative transition. (2) Nd3+ based NIR-II probes. The radiative energy transitions of 4F3/24I11/2 and 4F3/24I13/2 in Nd3+ enable the generation of intense NIR-II luminescence, peaked at 1060 and 1330~1340 nm, respectively. (3) NIR-II probes of other ions. In a similar radiative transition, NIR-II emission can also be realized from Ho3+ at 1155 nm (5I65I8), Pr3+ at 1289 nm (1G43H5), and Tm3+ at 1475 nm (3H43F4).
Figure 3. (a) Absorption (solid line) and NIR−II emission (solid line with color shade) spectra of different Ln3+. Adapted, with permission, from Ref. [36]. Copyright 2019, Wiley−VCH. (b) Energy level diagram and energy transfer of Ln3+ with NIR−II characteristic emission. Adapted, with permission, from Ref. [41]. Copyright 2019, Springer Nature. (c) NIR-II spectra of LnNCs with different contents of Ce3+ concentrations. Adapted, with permission, from Ref. [45]. Copyright 2019, American Chemical Society. (d) The energy transfer between Yb3+, Er3+, and Ce3+. Adapted, with permission, from Ref. [46]. Copyright 2017, Springer Nature. (e) NIR−II spectra of LnNCs with different Zn2+ concentrations. Adapted, with permission, from Ref. [47]. Copyright 2019, Springer Nature.
Figure 3. (a) Absorption (solid line) and NIR−II emission (solid line with color shade) spectra of different Ln3+. Adapted, with permission, from Ref. [36]. Copyright 2019, Wiley−VCH. (b) Energy level diagram and energy transfer of Ln3+ with NIR−II characteristic emission. Adapted, with permission, from Ref. [41]. Copyright 2019, Springer Nature. (c) NIR-II spectra of LnNCs with different contents of Ce3+ concentrations. Adapted, with permission, from Ref. [45]. Copyright 2019, American Chemical Society. (d) The energy transfer between Yb3+, Er3+, and Ce3+. Adapted, with permission, from Ref. [46]. Copyright 2017, Springer Nature. (e) NIR−II spectra of LnNCs with different Zn2+ concentrations. Adapted, with permission, from Ref. [47]. Copyright 2019, Springer Nature.
Chemosensors 10 00206 g003

2.2. Strategies of Optimizing NIR-II Luminescence

The LnNCs with good luminescent efficiency can be obtained by rationally selecting the matrix, sensitizing ion, and activating ion. However, the luminescent efficiency of LnNCs is still low due to the inherent problems of LnNCs, which severely limits its application in sensing [38,39,40,41,42]. It is possible to design and optimize the structure of LnNCs to increase the quantum yield and enhance its luminescent intensity.

2.2.1. Ion Doping

It is possible to increase the luminescent intensity of a specific wavelength by adjusting the energy distribution of Ln3+, in different luminescent bands, because of the multi-emission characteristics of Ln3+. The up-conversion luminescence of LnNCs can be converted into down-conversion luminescence by doping other ions into LnNCs, and the NIR-II luminescence can be enhanced [45,46,47]. Li et al. [45] synthesized NaLnF4: 40%Gd, 20%Yb, 2%Er, 5%Ce3+ nanorods (Ln = Y, Yb, Lu). The 1525 nm luminescence of Er3+ increased by 2.2 times, and the quantum efficiency reached 3.6% by doping Ce3+ to inhibit the up-conversion energy transfer of NaLnF4 (Figure 3c). The incorporation of Ce3+ can promote the relaxation process of 4I11/2 and 4I13/2 of Er3+, as well as shorten the life of the 4I11/2 state, thereby inhibiting the up-conversion process and enhancing Er3+ luminescence at 1525 nm (Figure 3d) [46]. However, high Ce3+ concentration (10%) will also cause concentration quenching and reduce the luminescence of NIR-II (Figure 3c).
In addition, the luminescent intensity of LnNCs involves 4f-4f electronic transition forbidden effect [46,47]. This prohibition can be broken by doping with other ions, resulting in luminescent enhancement of LnNCs. Zhong et al. [47] designed and synthesized a Zn-doped α-NaYbF4: 2%Er, 2%Ce, 10%Zn@NaYF4 NIR-II luminescent probe. The doping of Zn weakened the 4f-4f electronic transition forbidden effect, resulting in enhancing the NIR-II luminescence. When the doping concentration of Zn was 10%, the luminescence of NIR-II was 1.5 times higher than that of undoped α-ErNPs (Figure 3e).

2.2.2. Core-Shell Structure

Due to the small size of LnNCs, the excited state energy of its luminescent ions is easily transferred to the surface of the nanocrystals, through inter-ion energy transfer, and quenched by various surface defects, ligands, or solvent molecules. Therefore, coating different shell layers on mononuclear LnNCs can inhibit its surface quenching and cross-relaxation, thereby enhancing NIR-II luminescence.
At present, the main thing is to wrap an inert shell on the surface of a single core [48,49,50,51]. However, it should be noted that the lattice matching, between the inert shell layer and the core, is the main factor to be considered in the preparation. If the lattice parameter gap is too large, it is difficult to form a complete core-shell coating structure. Therefore, considering the factors of lattice matching, common inert shell layers include NaYF4, NaLuF4, and CaF2, etc. [49]. The luminescent intensity of the NaYF4: 10%Yb3+, 30%Nd3+@CaF2 core-shell structure at 1000 nm is 45 times higher than that of single-core NaYF4: 10%Yb3+, 30%Nd3+ under the excitation of 808 nm (Figure 4a) [48]. Likewise, Li et al. [49] completed a detailed investigation of the relationship between shell thickness and NIR-II emission intensity in the NaErF4@NaYF4 core/shell. As the inert shell path increases, the luminescence of Er3+ in NIR-II gradually increases (Figure 4b).
However, when the inert shell exceeds a certain thickness, it will weaken the light intensity received by the inner core, resulting in the weakening of the luminescent intensity. From Figure 4c, the luminescent intensity of LnNCs at 1525 nm was significantly reduced after NaYF4: Yb3+, Er3+ coated inert shell NaLuF4 with a thickness of 13.2 nm [50]. Sun et al. [51] reported a highly efficient NIR-II luminescent NaErF4@NaYF4@NaYF4: 10%Nd@NaYF4 nanocrystal. The strong NIR-II luminescence was successfully obtained by adjusting the thickness of the NaYF4 buffer layer, to limit the energy transfer between Er3+ and Nd3+, and optimizing the thickness of the outermost NaYF4 to passivate surface defects. Therefore, reasonable adjustment of the coating thickness of the inert shell layer and the position of the inert shell side can achieve high-efficiency luminescence in NIR-II.
In addition to wrapping the inert shell to achieve the enhancement of NIR-II luminescence, it can also wrap the active shell to enhance the NIR-II luminescence. The active shell layer refers to the presence of an activator or sensitizer in the shell layer, which mediates energy transfer and enhances the luminescence in the NIR-II [52]. Wang et al. [52] prepared β-NaGdF4@Na (Gd, Yb) F4: Er@NaYF4: Yb@NaNdF4: Yb. The Nd3+ in the outermost layer can be excited by an 800 nm laser, and the energy was transferred to Er3+ in the inner layer through Yb3+, resulting in an enhancement of its 1525 nm luminescence emission. Its 1525 nm luminescence penetrates the pork tissue to a depth of 18 mm.

2.2.3. Dye-Sensitized Luminescence

The absorption coefficient of Ln3+, in some specific wavelength bands, is very low, which greatly affects its luminescent intensity. The absorption coefficient of π-π transitions of some organic dyes is very high, has strong absorption in the spectrum 700~860 nm, and it can be used as a ligand to bind with Ln3+ [53,54,55,56]. The excited state energy level of the organic dye matches the excited state energy level of the Ln3+, which can produce the energy transfer process, so that the LnNCs can carry out efficient NIR-II luminescence emission (Figure 5a) [54].
Wang et al. [55] used indocyanine green dye (ICG) as an organic sensitizer to transfer energy to NaYF4: Er nanoparticles to enhance the NIR-II luminescence (Figure 5b,c). ICG, as a donor with high absorption cross-section at 808 nm, increased the excitation efficiency of Er3+ through the energy transfer mechanism (Figure 5d). However, the NIR-II luminescent intensity decreased when the concentration of ICG was higher, which was ascribed to the self-quenching effect between the ICG (Figure 5b). The luminescence at 1525 nm was enhanced by 10 times at the optimal ICG concentration. Similarly, Ren et al. [56] modified LnNCs with dye-brush polymer (Dye-BP), which led to an impressive 675 times enhancement of NIR-II luminescence in aqueous solution. When the concentration of Dye-BP exceeds a certain concentration, it will also reduce the luminescence of NIR-II. Therefore, it is the key to choosing suitable organic dyes as sensitizers, but it is necessary to find the optimal concentration of organic dyes in the regulation of enhancing NIR-II luminescence to achieve the strongest NIR-II luminescence.
In addition to organic sensitizers, inorganic nanoparticles are also used as sensitizers to enhance the NIR-II luminescence of LnNCs due to their wide absorption and narrow emission and excellent resistance to photobleaching. Zhang et al. [57] introduced an inorganic nanoparticle sensitized system, which uses near-infrared-emitting Ag2S QDs, as a sensitizer with broadband photon absorption, to enhance NIR-II luminescence. The NIR-II luminescence of LnNCs@Ag2S enhanced ~17-fold in intensity and ~10-fold in brightness over bare LnNCs because of increased absorptivity and overall broadening of the LnNCs@Ag2S absorption spectrum.
Except for the main methods mentioned above, it is also possible to enhance NIR-II luminescence by controlling the size and crystal form of synthetic LnNCs [47]. However, due to the inherent properties of LnNCs and the complexity of the external environment (-OH) during application, it will severely quench the luminescence in the NIR-II. Therefore, enhancing the NIR-II luminescence is a difficult point in research.

2.3. Preparation Methods

The grain size, crystal phase, morphology and shell of LnNCs are all key parameters that directly affect their luminescence performance. Researchers have been committed to developing various methods to synthesize LnNCs with different properties, such as thermal decomposition, hydrothermal synthesis, co-precipitation, and sol-gel process [42]. The most commonly used methods are thermal decomposition [58,59,60,61,62,63,64,65], hydrothermal synthesis [66,67,68,69,70,71] and co-precipitation [72,73,74,75,76,77,78,79,80,81].

2.3.1. Thermal Decomposition

Thermal decomposition is the most popular method to synthesize high-quality monodisperse LnNCs [58,59,60,61,62,63,64,65]. Thermal decomposition is a synthesis process in which organic precursors of metal ions are dissolved in a high-boiling organic solvent and then decomposed at high temperatures to obtain corresponding metal fluoride products.
The LnNCs with different crystal phases and sizes can be synthesized by controlling different reaction parameters. Suter et al. [59] studied the crystal phase process of synthesizing NaYF4 in oleic acid (OA) and octadecene by thermal decomposition method. There were three stages in this thermal decomposition synthesis process. The first stage was the nucleation and growth of small α-NaYF4 (<300 °C); the next stage was the growth of α-NaYF4 (300 °C, 60 min); the third stage was the phase transition from small α-NaYF4 to larger β-NaYF4 (300~310 °C, 30 min). Li et al. [60] also proved that α-NaYF4 can be synthesized at low temperature, while β-NaYF4 can be synthesized at high temperature, and the longer the time, the higher the crystallinity and the larger the particles. The content of OA in the reaction mainly affects the aspect ratio of the particles, and a high ratio of OA can synthesize a rod-like structure [60]. NH4F provides F ions in the process, which affects the nucleation rate of LnNCs, thus affecting the size of LnNCs, and the high NH4F content leads to the formation of small particles [58,60].
The LnNCs with different crystal types, sizes, and core-shell structures can be prepared using this method, especially for synthesizing ultra-small LnNCs. However, the requirements for the operation during the reaction and the amount of reactants added are particularly high when synthesizing small-sized materials, especially when the whole reaction is in an inert atmosphere. If oxygen enters, the OA oxidation solution will turn black and cause the reaction to fail. Wang et al. [58] studied, in detail, the problems and solutions that easily occur in synthesizing LnNCs via thermal decomposition. This has important guiding significance for the efficient synthesis of LnNCs.

2.3.2. Hydrothermal Synthesis

The hydrothermal synthesis usually occurs in a closed environment under high temperature and pressure. The high solubility and reactivity of the reactants in the sealed reaction vessel, under high temperature and high pressure, are used to grow nanocrystals in aqueous solutions (such as water or organic solvents) [42,66,67,68,69,70,71]. The reaction temperature is relatively low (generally less than 200 °C). It has become a simple and effective method to synthesize monodisperse LnNCs with adjustable morphology and structure [67,69,71].
Wang et al. [67] studied the effects of reactant concentration, temperature, and time on LnNCs. High concentration (Ln3+) would result in nanorods, while low concentration resulted in nanoparticles. Higher temperature will decrease the length-to-diameter ratio of the as-prepared nanorods. Too short of a reaction time resulted in only nanoparticles instead of nanorods. The morphology and size of LnNCs can also be controlled by controlling the amount of F. Qu et al. [69] used NH4F as the F source to prepare NaYF4 samples and found that different F/Y3+ ratios have a significant impact on the morphology, crystal phase, and size of the nanocrystals. The high molar ratio of F/Y3+ would benefit the α to β phase transition and formation of hexagonal phase products. The influence of different pH on the synthesis of LnNCs was also studied, NaYF4 presented an octahedral morphology and a smooth surface (pH 1.5). When the pH was 6, NaYF4 microtubules were formed [66].
The hydrothermal method can control the synthesis of LnNCs of different types of morphology, size, and crystal form, but the reaction rate of the system is slow, and it generally takes 20 h of reaction time to obtain monodisperse LnNCs with uniform size and morphology. The size is generally larger (sub-micron level). Meanwhile, it is difficult to observe the growth process of LnNCs. In addition, the operability of the hydrothermal reaction is slightly poor, so it is difficult to effectively control the growth of the core-shell structure.

2.3.3. Co-Precipitation

The co-precipitation method refers to the preparation of LnNCs with uniform composition in a variety of cationic solutions through a precipitation reaction after adding a precipitant [72,73,74,75,76,77,78,79,80,81]. Generally, the growth of LnNCs is controlled by adding ligands, such as polyvinylpyridinone (PVP), polyethyleneimine (PEI), or ethylenediaminetetraacetic acid (EDTA), to the solvent.
The co-precipitation method may be one of the most effective methods to prepare ultra-small LnNCs, and the size of LnNCs prepared by this method is uniform and controllable. Compared with other methods, this method does not need expensive equipment, the reaction conditions are relatively mild, the experimental operation process is relatively simple, and it saves time [75,78,79,80,81]. Yi et al. [78] improved this method and used simple water-soluble inorganic compounds as precursors to synthesize LaF3 nanocrystals, with uniform size distribution and size around 5 nm. Guan et al. [79] employed a facile co-precipitation method to synthesize the pure hexagonal NaYF4 and NaYF4: Yb, Er by controlling the molar ratio of anions to cations at room temperature and ambient pressure. They can realize the crystal phase transition of LnNCs by controlling the pH of the precursor liquid and obtaining the hexagonal phase in the pH range of 5.0~6.5. The whole process does not use any organic additives, nor high temperature and high pressure, and it is an environmentally friendly technology suitable for large-scale industrial production.
However, the morphology of LnNCs, obtained by the co-precipitation method, is relatively simple, the surface is rough, and the morphology is difficult to control. In addition, the luminescence of LnNCs is also relatively weak, and annealing is generally required to improve the crystallinity of the material and to improve the luminescent intensity. After the annealing treatment, the surfactant wrapped on the surface of the LnNCs will be carbonized, which will destroy the hydrophilicity of the LnNCs and affect its subsequent application.

3. Sensing Mechanisms of NIR-II LnNCs

The energy gap between some Stark sublevels of Ln3+ is very small, and the thermal coupling is very strong. Small temperature variations may result in remarkable changes in their emitting intensity [41,82,83,84], which are thus used for temperature sensing. Recently, Nexha et al. [85] gave a comprehensive review on LnNCs-based temperature sensing.
Aside from temperature, LnNCs have no direct relationship with any physiological and biochemical characteristics, due to their relatively stable structure and spectral characteristics. Therefore, in the construction of LnNCs-based bio/chemo sensors, LnNCs work either as energy donors or reference units, which further combine with an appropriate responsive unit to specifically respond to a target of interest. Some inorganic ions and organic dyes were used as the responsive units [44,51,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103], and typical LnNCs-based bio/chemo sensors are summarized in Table 1.

3.1. Inorganic-Ion-Based Recognition Mechanism

The LnNCs are internally doped or connected to inorganic ions on the surface, that are responsive to the external environment, to increase and decrease the luminescence of the NIR-II, thereby constructing a probe with a specific response ability to the external environment (Figure 6a). Inorganic ions must first participate in the energy transfer process of LnNCs NIR-II luminescence, and the increase and decrease in NIR-II luminescence can be controlled by the change of ion valence. Secondly, inorganic ions must respond to the external environment, and the change of the external environment will affect the change of the valence state of ions, as to realize the sensing function.
Lei et al. [86] designed and synthesized an H2O2 activated NaCeF4: Er, Yb NIR-II luminescent probe and realized the detection of uric acid (Figure 7a). Among them, the doped Ce3+ effectively enhanced the cross-relaxation process of 2F7/2 (Ce3+) + 4I11/2 (Er3+) → 2F5/2 (Ce3+) + 4I13/2 (Er3+) and greatly increased the number of excited state electrons of 4I13/2 (Er3+), thereby effectively inhibiting the up-conversion process and enhancing the down-conversion process, resulting in the nine-fold enhancement of the down conversion luminescence at 1530 nm. In this probe, Ce3+ existed as an external environmental responsive ion, which was used to construct a biosensing probe. Under the oxidation of H2O2, Ce3+ was oxidized to Ce4+, which weakened the cross-relaxation process and caused the luminescence quenching of the 1530 nm emission (Figure 7b). This responsive probe could be used to detect H2O2 or biomolecules that can generate H2O2. For example, the detection limit of uric acid could be reduced to 25.6 nmol·L−1 by detecting H2O2 produced by the reaction of uric acid and uricase (Figure 7c), and its tissue depth could reach 10 mm in small animal imaging. Similarly, Wang et al. [87] realized the response to H2S by attaching human serum albumin (HSA)-Ag+ (HSA-Ag+) to the surface of NaYF4: 20%Yb, 2%Er@NaYF4 (Figure 7d). In the presence of H2S, Ag2S QDs were formed in coated HSA through an H2S-induced chemical reaction between H2S and Ag+, which emitted luminescence at approximately 1050 nm (I808 Ex/1050) on irradiation with an 808 nm (Figure 7e). However, the luminescence signal of the probe was stable at 1550 nm (I980 Ex/1550) under 980 nm excitation (Figure 7f), generating a H2S concentration-dependent ratiometric I1050/1550 signal. Using this probe, the endogenous H2S could be real-time monitored, in vivo, by the ratio variation of luminescence signals in two channels of 1050 and 1550 nm.

3.2. Organic-Dye-Based Recognition Mechanism

The LnNCs are hardly individually used for bio/chemo sensing, due to their non-responsive property. However, detection pathways involving chemical reactions are the most common methods due to their high selectivity and sensitivity. To realize the NIR-II sensing of LnNCs, organic dyes can be introduced to the surface of the LnNCs nanoparticles.
The LnNCs themselves do not have the ability to respond to the external environment, and the ability to respond to the external environment is mainly accomplished by organic dyes. Organic dyes must affect the luminescence of LnNCs to change the responsive signal. On the one hand, they directly act on the luminescence of LnNCs to affect the luminescence of LnNCs. That is, organic dyes and LnNCs undergo energy transfer or luminous absorption (Figure 6b). On the other hand, organic dyes affect the luminescence of LnNCs by affecting the intensity of the excitation light source (Figure 6c).

4. Category of Sensing

4.1. Biomedical Related Species

The pathological microenvironment of diseased tissues is obviously different from the physiological environment of normal tissues, such as abnormal redox environment (ROS, HClO, ONOO, GSH, and H2S, etc.) [44,51,88,89,90,91,92,93,94,95,96,97,98,99,100,101]. These pathological parameters are exactly various diseases (such as cancer, inflammation, and cardiovascular disease) and essential biomarkers. According to the different response environments of LnNCs NIR-II probes, we have classified them.

4.1.1. Reactive Oxygen Species (ROS)

ROS including ·OH and H2O2, are widely present in organisms. It plays a vital role in physiological functions, which can regulate proteins, produce hormones, regulate cell signals, mediate inflammation, and eliminate pathogens [44,86,88,89,90].
Jia et al. [88] designed a cypate-modified NaErF4@NaLuF4 nanoprobe for detecting ·OH, on the basis of a typical reaction between cypate and·OH. Due to the presence of Er3+, NaErF4@NaLuF4 could produce 1550 nm NIR-II luminescence and 654 nm luminescence under excitation at 980 nm. The cypate strongly absorbed the luminescence at 654 nm, resulting in luminescence quenching at 654 nm, and it did not affect NIR-II emission at 1550 nm. When cypate encountered ·OH, the structure was destroyed, resulting in increased luminescence at 654 nm. Based on this, a proportional luminescent probe (I654/I1150) responding to ·OH was designed. The detection limit was 4.20 μM. In addition, the NaErF4@NaLuF4 nanoprobe was successfully used, in mice, for the diagnosis of arthritis in vivo. Similarly, different types of ratiometric ROS sensors have been constructed through different LnNCs and surface modifications of ROS-responsive organic dyes [44,89,90]. Among them, Liu et al. [44] used 1530 nm as the excitation light source to design an H2O2 sensor using up-conversion (I980/I1180), and Liao et al. [89,90] constructed the ROS sensor with dual excitation light sources (I980 Ex/I808 Ex). For the LnNCs NIR-II sensing of ROS, organic dyes are mainly used as response units, and the sensing is realized by affecting the NIR-II luminescence through organic dyes.

4.1.2. HClO

HClO is a weakly acidic reactive oxygen species, which has the physiological defense functions of antibacterial and anti-inflammatory. However, with the increase in HClO concentration, tissue damage and some diseases may occur, such as neuronal degeneration, arthritis, and so on [91,92,93,94,95].
Wang et al. [91] constructed a proportional luminescent probe, based on the Er3+ emission of 1550 nm, to detect the inflammatory site HOCl in vivo (Figure 8a). Here, the Cy7.5 fluorophore was chosen as the HOCl-responsive molecule. The Cy7.5 influenced the luminescence at 1550 nm through the selective absorption of different excitation light sources (I980 Ex/I808 Ex). Under 808 nm excitation, Cy7.5 had a stronger absorption at 808 nm, which greatly weakened the absorption of 808 nm by probe, resulting in weaker luminescence of Er3+ at 1530 nm. When HOCl molecules existed, Cy7.5 was degraded to weaken its absorption of 808 nm excitation. On the contrary, it enhanced the intensity of Er3+ NIR-II emission. Under 980 nm excitation, the luminescent intensity of Er3+ in 1550 nm remained almost unchanged, due to the weak absorption of Cy7.5 at 980 nm (Figure 8b). Therefore, under dual excitation wavelength irradiation, the ratio of the 1550 nm luminescent intensity of the two was closely related to the concentration of HOCl. The HOCl detection limit of the probe was 500 nmol/L, the resolution was 477 μm, and the detection depth was 3.5 mm in the living lymphoid inflammation model. Similarly, Zhang et al. [92] also designed a highly reactive oxygen species (HROS) sensor using dual excitation light sources (I980 Ex/I808 Ex). The detection limits for HOCl and ·OH were calculated as 0.3 and 1.6 μM, respectively. However, Cao et al. [93] reported a NIR-II ratio luminescent probe (I925 nm/I1525 nm) with a single excitation light source (I808 Ex) to detect HOCl by attaching organic dye Cy925 to the surface of NaYbF4: Er@NaYF4: Yb@NaYF4: Nd (Figure 8c). The response unit of HOCl is Cy925, which, itself, emits light at 925 nm. When Cy925 encounters HOCl, the structure of Cy925 will be destroyed, resulting in weakening of the fluorescence at 925 nm. The luminescence of Er3+, itself, at 1525 nm is not affected by HOCl. The detection range of the ratio probe for HOCl was 1~24 μM. The sensing of LnNCs NIR-II in HClO is similar to ROS, which is mainly realized by different organic dyes responding to HClO.

4.1.3. Glutathione (GSH)

GSH is an important endogenous antioxidant in the human body, which plays a vital role in the process of cell defense against toxins and free radical production. GSH has also been proven to be an important serum biochemical marker for the diagnosis of many diseases [96,97].
Wang et al. [96] reported a NIR-II nanoprobe consisting of 4-nitrophenol-Cy7 (NPh) conjugated LnNCs (NaYF4: 20%Yb, 2%Er@NaYF4: 30%Nd) for in vivo ratiometric sensing of GSH (Figure 8d). The GSH-responsive dye, NPh, has a strong absorption peak at 786 nm. Meanwhile, NPh has no fluorescence due to the presence of intramolecular photoinduced electron transfer (PET). In the presence of GSH, NPh reacted with GSH to generate Cy7-SG, the PET disappeared, and fluorescence recovered. Under 808 nm, Cy7-SG absorbed the energy of 808 nm, and transferred the excitation energy to LnNCs through energy transfer process, thereby achieving sensitization to LnNCs and enhanced luminescence at 1550 nm (Figure 8e). In addition, the 980 nm laser could not excite Cy7-SG, the luminescence at 1550 nm remained unchanged (Figure 8f), resulting in a ratio luminescence signal (I808 Ex/I980 Ex). The ratiometric I808 Ex/I980 Ex value exhibited a linear ship with GSH concentration, ranging from 0~24 mM, with a detection limit of 0.3 mM. This dual excitation light source can effectively realize the sensing of GSH. However, the different absorption of different excitation light sources in vivo may lead to different detection depths and limits in different tissues.

4.1.4. ONOO

ONOO is a very strong oxidant and plays an important role as an inflammatory mediator in physiology and pathology. ONOO and its secondary reactants can oxidize and destroy various biological molecules, such as proteins, lipids, and nucleic acids. This process has been implicated in cardiovascular disease, neurodegenerative diseases, host defense, and antitumor immune responses [51,98,99,104].
Zhao et al. [98] designed and synthesized the ONOO response NaYF4@NaYF4: 1%Nd3+@MY-1057 probe (Figure 9a). NaYF4@NaYF4: 1%Nd3+ absorbed 808 nm excitation energy and produced NIR-II luminescence at 1060 nm. MY-1057 molecules existed as response units that respond to ONOO and had a strong absorption at 1060 nm, thereby reducing the luminescent lifetime and intensity at 1060 nm. When ONOO existed, MY-1057 dye was degraded and the luminescent lifetime and intensity were restored (Figure 9b,c). The probe had no reference signal and was especially affected by tissue depth when using luminescent intensity for ONOO quantification. The slopes of intensity of the ONOO functions were 900, 432, and 60 a.u. µm−1 under 0, 2, and 5 mm penetration depth, respectively (Figure 9d). This illustrates the unreliable quantitative detection of ONOO using intensity imaging based on signal attenuation due to homogeneous scattering and absorption. In contrast, consistent lifetime response was obtained regardless of penetration depth. Under phantom tissue with various penetration depths, lifetime values exhibited linear correspondence to ONOO concentration, and the lifetime ONOO function slopes were almost identical (Figure 9e). When no reference signal was present, the luminescent lifetime was taken as the signal with better stability and penetration depth than the luminescent intensity as the signal. Therefore, using the luminescence lifetime of LnNCs NIR-II as a signal to realize sensing may be a better research direction.
Sun et al. [51] tactfully designed a ratio type NIR-II probe (Figure 9f) that was constructed by modifying organic dyes A1094, responsive to ONOO, on the surface of LnNCs (NaErF4@NaYF4@NaYF4: 10%Nd@NaYF4). The LnNCs exhibited two NIR-II luminescence emissions of 1525 nm from Er3+ and 1060 nm from Nd3+. Due to the substantial overlap between the A1094 absorbance at 1094 nm and the LnNCs@A1094 emission at 1060 nm, the 1060 nm emission was quenched by A1094 via the efficient Förster resonance energy transfer. The ONOO is a highly specific oxidant of A1094, which can destroy its absorbance at 1094 nm. Therefore, the emission at 1060 nm of LnNCs@A1094 could be activated in the presence of ONOO (Figure 9g). Meanwhile, A1094 had no effect on the luminescence at 1525 nm of LnNCs@A1094 (Figure 9h). The ratiometric luminescent intensity (I1060/I1525) of LnNCs@A1094 increased, linearly, with the augment of ONOO concentration in the range of 0~6 μM, and the detection limit was 0.8 μM (Figure 9i).

4.1.5. H2S

H2S is the third gas signal molecule discovered after NO and CO. It plays an important role in the pathogenesis of neurodegenerative diseases, diabetes, heart failure, inflammation, cancer, and other diseases [87,100,101].
Liu et al. [100] developed a NIR-II luminescent LnNCs (NaGdF4:2%Nd@NaGdF4) probe for sensitive and selective sensing of H2S. In this probe, the H2S response unit is compound 1, which has high absorption at 808 nm (Figure 10a). Due to the weakening of the luminescent intensity of 808 nm excitation, the NIR-II emission at 1060 nm (Nd3+) was weakened (Figure 10b). After the reaction with H2S, the nucleophilic addition reaction between H2S and the benzpyrole group in compound 1 could bleach the absorption of compound 1 at 808 nm and recover the 1060 nm emission of LnNCs (Figure 10c). The luminescent intensity at 1060 nm exhibited a good linear relationship in the range from 5 to 750 μM, and the detection limit was 17 nM.
Compared with NIR-II probes that use organic dyes to achieve H2S response, Deng et al. [101] achieved H2S responsive NIR-II ratio probes by coating LnNCs (NaYF4: Gd, Yb, Er@NaYF4: Yb) with SiO2 and Ag nanodots (Figure 10d). Here, Ag nanodots were used as the signal unit of H2S response. After sulfidation with S2−, LnNCs@SiO2@Ag was quickly converted to LnNCs@SiO2@Ag2S. Under 980 nm laser, the luminescence of LnNCs@SiO2@Ag and LnNCs@SiO2@Ag2S at 1525 nm were basically unchanged. It did not change with the increase in the amount of H2S, which could be used as a reference signal (Figure 10e,f). Under 808 nm excitation, LnNCs@SiO2@Ag has no luminescence emission at 1053 nm. However, when Ag2S was formed, LnNCs@SiO2@Ag2S had a significant luminescence emission at 1053 nm under excitation at 808 nm (Figure 10e,g). Using the detection (I808 Ex/1053 Em) and reference (I980 Ex/1525 Em) signals, the NIR-II ratiometric luminescence signal, presented linearly, increases via improving the content of Na2S from 0 to 0.08 μM, and the detection limit was 0.7 nM. More importantly, the in situ highly specific ratiometric imaging of the metformin-induced hepatotoxicity was successfully achieved by using the activatable orthogonal NIR-II emitting probe. Compared with organic dyes as the response unit, Ag, as the response unit, has better responsiveness and detection limit for H2S. The main reason for this may be that the Ag has better stability and is less prone to falling off.

4.2. Chemical Species

In vivo tracking of drug carriers and monitoring of drug release processes are of great significance for evaluating drug delivery efficiency and pharmacokinetics. Wang et al. [102] proposed a competitive absorption strategy based on the multi-wavelength excitation characteristics of LnNCs (NaGdF4: 5%Nd@NaGdF4), and designed a micron-sized oral drug carrier that could monitor the release process of protein drugs in the intestine (Figure 11a). They coated LnNCs in a shell of silica. Then, its surface was wrapped a layer of mesoporous silica to form SiO2-Nd@SiO2@mSiO2-NH2 (mSiO2-Nd) (Figure 11b). Protein drugs (BSA) and tetrasulfonated phthalocyanine sodium salt (NPTAT) will form BSA-NPTAT complexes due to electrostatic interactions. The high absorption coefficient of the NPTAT, at 730 nm, could dramatically quench the 1060 nm luminescence of mSiO2-Nd when excited by 730 nm (Figure 11c,d). While under 808 nm excitation, the mSiO2-Nd 1060 nm emission was barely influenced by the NPTAT due to the weak absorbance of NPTAT at 808 nm. When BSA-NPTAT was loaded into mSiO2-Nd, the release process of the drug was monitored and quantified by the emission at 1060 nm under the dual excitation light source (I730/I808). With the gradual release of BSA-NPTAT, the luminescence of Nd3+, under excitation at 730 nm, also gradually recovered. The ratio of I730/I808 reflected the degree of drug release (Figure 11e).
The LnNCs NIR-II luminescence applications in chemical sensing are seldom compared to the broad applications in biosensing. The main reason is that the biggest bright spot of the current NIR-II luminescence, compared with the luminescence of other regions, is the deeper tissue penetration and lower biological background interference. Meanwhile, NIR-II luminescence requires a specific and expensive NIR-II luminous receiver. Therefore, the application of NIR-II chemical sensing can only be based on the distribution of pharmaceutical chemicals or the concentration of chemical substances in organisms. Recently, Su et al. [103] developed an LnNCs NIR-II probe to detect inorganic pyrophosphate (PPi) in vivo for the first time. However, there are very few reports on the NIR-II sensing of LnNCs for chemical substances in vivo, which is also a direction for future sensing research.

5. Conclusions and Outlook

In this paper, NIR-II luminescent LnNCs-based bio/chemo sensors are reviewed. Firstly, the luminescence mechanism of LnNCs is briefly presented, followed by the enhancement strategies of their NIR-II luminescence (ion doping, core-shell structure, and dye sensitization) and preparing methods (thermal decomposition, hydrothermal synthesis, and co-deposition method). We then comprehensively review the sensing mechanism of the LnNCs-based bio/chemo sensors, as well as their applications in detection of various chemical and biomedical-related species. Obviously, NIR-II LnNCs-based bio/chemo sensors show a great prospect in biomedical sensing and imaging. However, they also confront some practical problems when working under physiological environments.
One is the luminescence quenching of NIR-II LnNCs by ambient hydroxyl groups in aqueous medium [2,37]. Although core-shell structure has been proved to be an effective strategy to isolate Ln3+ ions from external environment, the non-radiative decay by hydroxyl group is very efficient. This put forwards strict requirements for shell coating, which should not only be thick enough to shield the vibrational decaying of nearby hydroxyl groups but should also be compact enough to prevent the infiltrating of H2O molecules. On the other hand, the quantum yield of LnNCs is not high (<30%) due to its structure and lattice defects [12,36]. Higher luminescence can be achieved by structural design, doping ions to fill emission levels, or building shell structures in nanoparticles, but this also creates high requirements for synthesis conditions, which is not conducive to large-scale production and subsequent applications. The LnNCs-based bio/chemo sensors show rather weak luminescent intensity because of the inherent small absorption cross-section of Ln3+ ions.
In addition, organic dyes are commonly used as fluorescent probes in LnNCs-based bio/chemo sensors [36]. The coupling between organic dyes and organic LnNCs needs to be considered in case of dye leakage, which may reduce the sensing accuracy. It is well known that most of these emissions lie in the visible range, which would degrade the performance of NIR-II sensors with shallow penetration depth. Hence, it is urgent to develop more fluorescent organic probes with NIR emission to match the strong penetrability of NIR-II LnNCs. Meanwhile, current research shows that most LnNPs are harmless luminescent probes suitable for living cells and mice [38,39,105]. However, the application of LnNPs in the body system is still limited due to the complex environment and metabolic processes in the body. There is an urgent need for a systematic and comprehensive assessment of their toxicity and stability in cells and animals.
Last but not least, NIR-II luminescent LnNCs-based bio/chemo sensors exhibit high sensitivity by virtue of deep penetration and background-free detection, but this is at the cost of expensive detectors, such as InGaAs [41,42,105]. This severely hampers their widespread applications in the biomedical realm, particularly in the case of lifetime-based detection. NIR-II LnNCs-based bio/chemo sensors may witness broader applications with the further development of NIR photodetectors.

Author Contributions

Conceptualization, T.Y., J.Q. and H.P.; methodology, T.Y., J.Q. and J.Z.; software, J.Z. and L.G.; validation, T.Y., J.Z. and L.G.; formal analysis, X.W. and M.Y. (Mei You); investigation, J.Z., M.Y. (Mu Yang) and L.G.; resources, H.P.; data curation, T.Y., J.Q. and H.P.; writing—original draft preparation, T.Y.; writing—review and editing, T.Y., J.Q. and H.P.; visualization, T.Y., L.G. and H.P.; supervision, M.Y. (Mu Yang) and H.P.; project administration, H.P.; funding acquisition, H.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work by the National Natural Science Foundation of China (Grant Nos.62175266, 61775245) and the Interdisciplinary Research Project of MUC (2020MDJC10).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We gratefully acknowledge support of this work by the National Natural Science Foundation of China (Grant Nos.62175266, 61775245) and the Interdisciplinary Research Project of MUC (2020MDJC10).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Liu, Y.; Li, Y.; Koo, S.; Sun, Y.; Liu, Y.; Liu, X.; Pan, Y.; Zhang, Z.; Du, M.; Lu, S.; et al. Versatile types of inorganic/organic NIR-IIa/IIb fluorophores: From strategic design toward molecular imaging and theranostics. Chem. Rev. 2022, 122, 209–268. [Google Scholar] [CrossRef] [PubMed]
  2. Zhang, S.T.; Wang, Y.H.; Zhang, H.J. Lanthanide-doped fluorescence probes for NIR-II fluorescence imaging. Chin. J. Lumin. 2020, 41, 1460–1478. [Google Scholar] [CrossRef]
  3. Bhat, M.P.; Kurkuri, M.; Losic, D.; Kigga, M.; Altalhi, T. New optofluidic based lab-on-a-chip device for the real-time fluoride analysis. Anal. Chim. Acta 2021, 1159, 338439. [Google Scholar] [CrossRef] [PubMed]
  4. Purves, R.W.; Souster, K.; West, M.; Huda, A.M.; Fisher, C.M.E.; Belford, M.W.; Shurmer, B.O. Improved thyreostatic drug detection in animal tissues using liquid chromatography-high-field asymmetric waveform ion mobility spectrometry-mass spectrometry. J. Agric. Food Chem. 2022, 70, 4785–4791. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, J. Electrochemical detection for microscale analytical systems: A review. Talanta 2002, 56, 223–231. [Google Scholar] [CrossRef]
  6. An, X.; Zhu, X.; Liu, J.; Zou, L.; Li, G.; Ye, B. Ratiometric fluorescence detection of ciprofloxacin using the terbium-based coordination polymers. Spectrochim. Acta A 2022, 269, 120775. [Google Scholar] [CrossRef]
  7. Liu, S.; Yan, L.; Huang, J.; Zhang, J.; Zhou, B. Controlling upconversion in emerging multilayer core-shell nanostructures: From fundamentals to frontier applications. Chem. Soc. Rev. 2022, 51, 1729–1765. [Google Scholar] [CrossRef]
  8. Cai, L.; Wang, Z.; Lin, B.; Liu, K.; Wang, Y.; Yuan, Y.; Tao, X.; Lv, R. Rare earth nanoparticles for sprayed and intravenous NIR II imaging and photodynamic therapy of tongue cancer. Nanoscale Adv. 2022, 4, 2224–2232. [Google Scholar] [CrossRef]
  9. Tao, W.; Farokhzad, O.C. Theranostic nanomedicine in the NIR-II window: Classification, fabrication, and biomedical applications. Chem. Rev. 2022, 122, 5405–5407. [Google Scholar] [CrossRef]
  10. Liang, T.; Guo, Z.; He, Y.; Wang, Y.; Li, C.; Li, Z.; Liu, Z. Cyanine-doped lanthanide metal-organic frameworks for near-Infrared II bioimaging. Adv. Sci. 2022, 9, 2104561. [Google Scholar] [CrossRef]
  11. Zhao, M.Y.; Li, B.H.; Zhang, H.X.; Zhang, F. Activatable fluorescence sensors for in vivo bio-detection in the second near-infrared window. Chem. Sci. 2021, 12, 3448. [Google Scholar] [CrossRef] [PubMed]
  12. Ge, X.Q.; Wei, R.Y.; Sun, L.N. Lanthanide nanoparticles with efficient near-infrared-II emission for biological applications. J. Mater. Chem. B 2020, 8, 10257. [Google Scholar] [CrossRef] [PubMed]
  13. Diao, S.; Hong, G.S.; Antaris, A.L.; Blackburn, J.L.; Cheng, K.; Cheng, Z.; Dai, H.J. Biological imaging without autofluorescence in the second near-infrared region. Nano Res. 2015, 8, 3027–3034. [Google Scholar] [CrossRef]
  14. Hong, G.S.; Antaris, A.L.; Dai, H.J. Near-infrared fluorophores for biomedical imaging. Nat. Biomed. Eng. 2017, 1, 0010. [Google Scholar] [CrossRef]
  15. Naczynski, D.J.; Tan, M.C.; Zevon, M.; Wall, B.; Kohl, J.; Kulesa, A.; Chen, S.; Roth, C.M.; Riman, R.E.; Moghe, P.V. Rare-earth-doped biological composites as in vivo shortwave infrared reporters. Nat. Commun. 2013, 4, 2199. [Google Scholar] [CrossRef]
  16. Tsukasaki, Y.; Komatsuzaki, A.; Mori, Y.; Ma, Q.; Yoshioka, Y.; Jin, T. A short-wavelength infrared emitting multimodal probe for non-invasive visualization of phagocyte cell migration in living mice. Chem. Commun. 2014, 50, 14356–14359. [Google Scholar] [CrossRef]
  17. Lei, Z.H.; Zhang, F. Molecular engineering of NIR-II fluorophores for improved biomedical detection. Angew. Chem. Int. Ed. 2021, 133, 16430–16444. [Google Scholar] [CrossRef]
  18. Zhong, Y.; Dai, H.J. A mini-review on rare-earth down-conversion nanoparticles for NIR-II imaging of biological systems. Nano Res. 2020, 13, 1281–1294. [Google Scholar] [CrossRef]
  19. Tang, Y.F.; Pei, F.; Lu, X.M.; Fan, Q.L.; Huang, W. Recent advances on activatable NIR-II fluorescence probes for biomedical imaging. Adv. Opt. Mater. 2019, 7, 1900917. [Google Scholar] [CrossRef]
  20. Kong, Y.; Chen, J.; Fang, H.; Heath, G.; Wo, Y.; Wang, W.; Li, Y.; Guo, Y.; Evans, S.D.; Chen, S.; et al. Highly fluorescent ribonuclease-A-encapsulated lead sulfide quantum dots for ultrasensitive fluorescence in vivo imaging in the second near-infrared window. Chem. Mater. 2016, 28, 3041–3050. [Google Scholar] [CrossRef] [Green Version]
  21. Yang, T.; Tang, Y.; Ling Liu, L.; Lv, X.Y.; Wang, Q.L.; Ke, H.; Deng, Y.B.; Yang, H.; Yang, X.; Liu, G.; et al. Size-dependent Ag2S nanodots for second near-infrared fluorescence/photoacoustics imaging and simultaneous photothermal therapy. ACS Nano 2017, 11, 1848–1857. [Google Scholar] [CrossRef] [PubMed]
  22. Robinson, J.T.; Hong, G.; Liang, Y.; Zhang, B.; Yaghi, O.K.; Dai, H.J. In vivo fluorescence imaging in the second near-infrared window with long circulating carbon nanotubes capable of ultrahigh tumor uptake. J. Am. Chem. Soc. 2012, 134, 10664–10669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wan, H.; Yue, J.; Zhu, S.; Uno, T.; Zhang, X.; Yang, Q.; Yu, K.; Hong, G.; Wang, J.; Li, L.; et al. A bright organic NIR-II nanofluorophore for three-dimensional imaging into biological tissues. Nat. Commun. 2018, 9, 1171. [Google Scholar] [CrossRef] [PubMed]
  24. Dang, H.P.; Yan, L.F. Organic fluorescent nanoparticles with NIR-II emission for bioimaging and therapy. Biomed. Mater. 2021, 16, 022001. [Google Scholar] [CrossRef]
  25. Wang, S.F.; Li, B.H.; Zhang, F. Molecular fluorophores for deep-tissue bioimaging. ACS Cent. Sci. 2020, 6, 1302–1316. [Google Scholar] [CrossRef]
  26. Sun, W.; Guo, S.; Hu, C.; Fan, J.; Peng, X. Recent Development of chemosensors based on cyanine platforms. Chem. Rev. 2016, 116, 7768–7817. [Google Scholar] [CrossRef]
  27. Wanderi, K.; Cui, Z.Q. Organic fluorescent nanoprobes with NIR-IIb characteristics for deep learning. Exploration 2022, 2, 20210097. [Google Scholar] [CrossRef]
  28. Zhou, H.J.; Ren, T.B. Recent progress of cyanine fluorophores for NIR-II sensing and imaging. Chem. Asian J. 2022, 17, e202200147. [Google Scholar] [CrossRef]
  29. Zhang, X.; Li, S.; Ma, H.; Wang, H.; Zhang, R.; Zhang, X.D. Activatable NIR-II organic fluorescent probes for bioimaging. Theranostics 2022, 12, 3345–3371. [Google Scholar] [CrossRef]
  30. Yu, Z.; He, Y.; Schomann, T.; Wu, K.; Hao, Y.; Suidgeest, E.; Zhang, H.; Eich, C.; Cruz, L.J. Achieving effective multimodal imaging with rare-earth ion-doped CaF2 nanoparticles. Pharmaceutics 2022, 14, 840. [Google Scholar] [CrossRef]
  31. Xu, H.; Yang, Y.; Lu, L.; Yang, Y.; Zhang, Z.; Zhao, C.X.; Zhang, F.; Fan, Y. Orthogonal multiplexed NIR-II imaging with excitation-selective lanthanide-based nanoparticles. Anal. Chem. 2022, 94, 3661–3668. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, Q.X.; Yang, Y.F.; Yang, X.F.; Pan, Y.; Sun, L.D.; Zhang, W.Y.; Shao, Y.; Shen, J.; Lin, J.; Li, L.; et al. Upconverted/downshifted NaLnF4 and metal-organic framework heterostructures boosting NIR-II imaging-guided photodynamic immunotherapy toward tumors. Nano Today 2022, 43, 101439. [Google Scholar] [CrossRef]
  33. Lv, Z.; Jin, L.; Cao, Y.; Zhang, H.; Xue, D.; Yin, N.; Zhang, T.; Wang, Y.; Liu, J.; Liu, X.; et al. A nanotheranostic agent based on Nd3+-doped YVO4 with blood-brain-barrier permeability for NIR-II fluorescence imaging/magnetic resonance imaging and boosted sonodynamic therapy of orthotopic glioma. Light Sci Appl 2022, 11, 116. [Google Scholar] [CrossRef] [PubMed]
  34. Gu, Y.; Guo, Z.; Yuan, W.; Kong, M.; Liu, Y.; Liu, Y.T.; Gao, Y.; Feng, W.; Wang, F.; Zhou, J.; et al. High-sensitivity imaging of time-domain near-infrared light transducer. Nat. Photonics 2019, 13, 525–531. [Google Scholar] [CrossRef]
  35. Ding, S.W.; Lu, L.F.; Fan, Y.; Zhang, F. Recent progress in NIR-Ⅱ emitting lanthanide-based nanoparticles and their biological applications. J. Rare Earths 2020, 38, 451–463. [Google Scholar] [CrossRef]
  36. Fan, Y.; Zhang, F. A new generation of NIR-Ⅱ probes: Lanthanide-based nanocrystals for bioimaging and biosensing. Adv. Opt. Mater. 2019, 7, 1801417. [Google Scholar] [CrossRef]
  37. Wang, Z.; Xing, B.G. Near-infrared multipurpose lanthanide-imaging nanoprobes. Chem.-Asian J. 2020, 15, 2076–2091. [Google Scholar] [CrossRef]
  38. Xu, J.; Gulzar, A.; Yang, P.P.; Bi, H.; Yang, D.; Gai, S.; He, F.; Lin, J.; Xing, B.G.; Jin, D.Y. Recent advances in near-infrared emitting lanthanide-doped nanoconstructs: Mechanism, design and application for bioimaging. Coord. Chem. Rev. 2019, 381, 104–134. [Google Scholar] [CrossRef]
  39. Yang, Y.J.; Tu, D.; Zhang, Y.; Zhang, P.; Chen, X.Y. Recent advances in design of lanthanide-containing NIR-II luminescent nanoprobes. iScience 2021, 24, 102062. [Google Scholar] [CrossRef]
  40. Zhang, M.Z.; Zhai, X.Y.; Sun, M.Z.; Ma, T.F.; Huang, Y.K.; Huang, B.L.; Du, Y.P.; Yan, C.H. When rare earth meets carbon nanodots: Mechanisms, applications and outlook. Chem. Soc. Rev. 2020, 49, 9220–9248. [Google Scholar] [CrossRef]
  41. Yu, S.H.; Tu, D.; Lian, W.; Xu, J.; Chen, X.Y. Lanthanide-doped near-infrared II luminescent nanoprobes for bioapplications. Sci. China Mater. 2019, 62, 1071–1086. [Google Scholar] [CrossRef] [Green Version]
  42. Lia, Z.; Ding, X.; Cong, H.; Wang, S.; Yu, B.; Shen, Y. Recent advances on inorganic lanthanide-doped NIR-II fluorescence nanoprobes for bioapplication. J. Lumin. 2020, 228, 117627. [Google Scholar] [CrossRef]
  43. Zhang, H.X.; Fan, Y.; Pei, P.; Sun, C.; Lu, L.; Zhang, F. Tm3+-sensitized NIR-II fluorescent nanocrystals for in vivo information storage and decoding. Angew. Chem. Int. Ed. 2019, 58, 10153–10157. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, L.; Wang, S.F.; Zhao, B.Z.; Pei, P.; Fan, Y.; Li, X.M.; Zhang, F. Er3+ sensitized 1530 nm to 1180 nm second near-infrared window upconversion nanocrystals for in vivo biosensing. Angew. Chem. Int. Ed. 2018, 57, 7518–7522. [Google Scholar] [CrossRef]
  45. Li, Y.B.; Zeng, S.J.; Hao, J.H. Non-invasive optical guided tumor metastasis/vessel imaging by using lanthanide nanoprobe with enhanced down-shifting emission beyond 1500 nm. ACS Nano 2019, 13, 248–259. [Google Scholar] [CrossRef]
  46. Zhong, Y.; Ma, Z.; Zhu, S.; Yue, J.; Zhang, M.; Antaris, A.L.; Yuan, J.; Cui, R.; Wan, H.; Zhou, Y.; et al. Boosting the down-shifting luminescence of rare-earth nanocrystals for biological imaging beyond 1500 nm. Nat. Commun. 2017, 8, 737. [Google Scholar] [CrossRef] [Green Version]
  47. Zhong, Y.; Ma, Z.; Wang, F.; Wang, X.; Yang, Y.; Liu, Y.; Zhao, X.; Li, J.; Du, H.; Zhang, M.; et al. In vivo molecular imaging for immunotherapy using ultra-bright near-infrared-Ⅱb rare-earth nanoparticles. Nat. Biotechnol. 2019, 37, 1322–1331. [Google Scholar] [CrossRef]
  48. Tan, M.; Rosal, B.; Zhang, Y.; Rodríguez, E.M.; Hu, J.; Zhou, Z.; Fan, R.; Ortgies, D.H.; Fernández, N.; Chaves-Coira, I.; et al. Rare-earth-doped fluoride nanoparticles with engineered long luminescence lifetime for time-gated in vivo optical imaging in the second biological window. Nanoscale 2018, 10, 17771–17780. [Google Scholar] [CrossRef] [Green Version]
  49. Li, H.; Wang, X.; Li, X.L.; Zeng, S.J.; Chen, G.Y. Clearable shortwave-infrared-emitting NaErF4 nanoparticles for noninvasive dynamic vascular imaging. Chem. Mater. 2020, 32, 3365–3375. [Google Scholar] [CrossRef]
  50. Fischer, S.; Bronstein, N.D.; Swabeck, J.K.; Chan, E.M.; Alivisatos, A.P. Precise tuning of surface quenching for luminescence enhancement in core-shell lanthanide-doped nanocrystals. Nano Lett. 2016, 16, 7241–7247. [Google Scholar] [CrossRef] [Green Version]
  51. Sun, Z.; Huang, H.; Zhang, R.; Yang, X.; Yang, H.; Li, C.; Zhang, Y.; Wang, Q.B. Activatable rare earth near-infrared-II fluorescence ratiometric nanoprobes. Nano Lett. 2021, 21, 6576–6583. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, R.; Li, X.M.; Zhou, L.; Zhang, F. Epitaxial seeded growth of rare-earth nanocrystals with efficient 800 nm near-infrared to 1525 nm short-wavelength infrared down conversion photoluminescence for in vivo bioimaging. Angew. Chem. Int. Ed. 2015, 126, 12282–12286. [Google Scholar] [CrossRef]
  53. Wang, Q.; Liang, T.; Wu, J.; Li, Z.; Liu, Z.H. Dye-sensitized rare earth-doped nanoparticles with boosted NIR-IIb emission for dynamic imaging of vascular network-related disorders. ACS Appl. Mater. Interfaces 2021, 13, 29303–29312. [Google Scholar] [CrossRef]
  54. Shao, W.; Chen, G.Y.; Kuzmin, A.; Kutscher, H.L.; Pliss, A.; Ohulchanskyy, T.Y.; Prasad, P.N. Tunable narrow band emissions from dye-sensitized core/shell/shell nanocrystals in the second near-infrared biological window. J. Am. Chem. Soc. 2016, 138, 16192–16195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wang, D.; Wang, D.; Kuzmin, A.; Pliss, A.; Shao, W.; Xia, J.; Qu, J.; Prasad, P.N. ICG-sensitized NaYF4:Er nanostructure for theranostics. Adv. Optical Mater. 2018, 6, 1701142. [Google Scholar] [CrossRef]
  56. Ren, F.; Liu, H.; Zhang, H.; Jiang, Z.; Xia, B.; Genevois, C.; He, T.; Allix, M.; Sun, Q.; Li, Z.; et al. Engineering NIR-IIb fluorescence of Er-based lanthanide nanoparticles for through-skull targeted imaging and imaging-guided surgery of orthotopic glioma. Nano Today 2020, 34, 100905. [Google Scholar] [CrossRef]
  57. Zhang, W.; Chen, T.; Su, L.; Ge, X.; Chen, X.Y.; Song, J.B.; Yang, H.H. Quantum dot-based sensitization system for boosted photon absorption and enhanced second near-infrared luminescence of lanthanide-doped nanoparticle. Anal. Chem. 2020, 92, 6094–6102. [Google Scholar] [CrossRef]
  58. Wang, F.; Deng, R.R.; Liu, X.G. Preparation of core-shell NaGdF4 nanoparticles doped with luminescent lanthanide ions to be used as upconversion-based probes. Nat. Protoc. 2014, 9, 1634–1644. [Google Scholar] [CrossRef]
  59. Suter II, J.D.; Pekas, N.J.; Berry, M.T.; May, P.S. Real-time-monitoring of the synthesis of β-NaYF4:17% Yb, 3% Er nanocrystals using NIR-to-visible upconversion luminescence. J. Phys. Chem. C 2014, 118, 13238–13247. [Google Scholar] [CrossRef]
  60. Li, D.D.; Shao, Q.Y.; Dong, Y.; Jiang, J.Q. Phase-, shape- and size-controlled synthesis of NaYF4:Yb3+,Er3+ nanoparticles using rare-earth acetate precursors. J. Rare Earth. 2014, 32, 1032–1036. [Google Scholar] [CrossRef]
  61. Chen, B.; Kong, W.; Wang, N.; Zhu, G.Y.; Wang, F. Oleylamine-mediated synthesis of small NaYbF4 nanoparticles with tunable size. Chem. Mater. 2019, 31, 4779–4786. [Google Scholar] [CrossRef]
  62. Rastogi, C.K.; Lu, E.; Tam, J.; Pichaandi, J.M.; Howe, J.; Winnik, M.A. Influence of the sodium precursor on the cubic-to-hexagonal phase transformation and controlled preparation of uniform NaNdF4 nanoparticles. Langmuir 2021, 37, 2146–2152. [Google Scholar] [CrossRef] [PubMed]
  63. Huang, P.; Zheng, W.; Gong, Z.; You, W.; Wei, J.; Chen, X. Rare earth ion- and transition metal ion-doped inorganic luminescent nanocrystals: From fundamentals to biodetection. Mater. Today Nano 2019, 5, 100031. [Google Scholar] [CrossRef]
  64. You, W.; Tu, D.; Zheng, W.; Shang, X.; Song, X.; Zhou, S.; Liu, Y.; Li, R.; Chen, X. Large-scale synthesis of uniform lanthanide-doped NaREF4 upconversion/ downshifting nanoprobes for bioapplications. Nanoscale 2018, 10, 11477–11484. [Google Scholar] [CrossRef] [PubMed]
  65. Cheng, X.; Ge, H.; Wei, Y.; Zhang, K.; Su, W.; Zhou, J.; Yin, L.; Zhan, Q.; Jing, S.; Huang, L. Design for brighter photon upconversion emissions via energy level overlap of lanthanide ions. ACS Nano 2018, 12, 10992–10999. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, Y.; Gai, S.; Niu, N.; He, F.; Yang, P.P. Synthesis of NaYF4 microcrystals with different morphologies and enhanced up-conversion luminescence properties. Phys. Chem. Chem. Phys. 2013, 15, 16795. [Google Scholar] [CrossRef]
  67. Wang, L.; Li, Y.D. Controlled synthesis and luminescence of lanthanide doped NaYF4 nanocrystals. Chem. Mater. 2007, 19, 727–734. [Google Scholar] [CrossRef]
  68. Sun, L.; Wei, R.; Feng, J.; Zhang, H.J. Tailored lanthanide-doped upconversion nanoparticles and their promising bioapplication prospects. Coordin. Chem. Rev. 2018, 364, 10–32. [Google Scholar] [CrossRef]
  69. Qu, X.; Pan, G.; Yang, H.K.; Chen, Y.; Chung, J.W.; Moon, B.K.; Choi, B.C.; Jeong, J.H. Solvothermal synthesis and luminescence properties of NaYF4:Ln3+ (Eu3+, Tb3+, Yb3+/Er3+) nano- and microstructures. Opt. Mater. 2012, 34, 1007–1012. [Google Scholar] [CrossRef]
  70. Zhou, Z.; Zheng, W.; Kong, J.; Liu, Y.; Huang, P.; Zhou, S.; Chen, Z.; Shi, J.; Chen, X. Rechargeable and LED-activated ZnGa2O4: Cr3+ near-infrared persistent luminescence nanoprobes for background-free biodetection. Nanoscale 2017, 9, 6846–6853. [Google Scholar] [CrossRef]
  71. Cao, T.M.D.; Le, T.T.G.; Nguyen, T.P.N.; Dau, T.A.N.; Nguyen, V.T.; Tran, T.T.V. Investigating the effect of Yb3+ and Er3+ concentration on red/green luminescent ratio in β-NaYF4: Er, Yb nanocrystals using spectroscopic techniques. J. Mol. Struct. 2020, 12, 128014. [Google Scholar] [CrossRef]
  72. Carniato, F.; Thangavel, K.; Tei, L.; Botta, M. Structure and dynamics of the hydration shells of citrate-coated GdF3 nanoparticles. J. Mater. Chem. B 2013, 1, 2442. [Google Scholar] [CrossRef] [PubMed]
  73. Evanics, F.; Diamente, P.R.; van Veggel, F.C.J.M.; Stanisz, G.J.; Prosser, R.S. Water-soluble GdF3 and GdF3/LaF3 nanoparticless-physical characterization and NMR relaxation properties. Chem. Mater. 2006, 18, 2499. [Google Scholar] [CrossRef]
  74. Bednarkiewicz, A.; Mech, A.; Karbowiak, M.; Strek, W. Spectral properties of Eu3+ doped NaGdF4 nanocrystals. J. Lumin. 2005, 114, 247. [Google Scholar] [CrossRef]
  75. Loo, J.F.C.; Chien, Y.H.; Yin, F.; Kong, S.K.; Ho, H.P.; Yong, K.T. Upconversion and downconversion nanoparticles for biophotonics and nanomedicine. Coord. Chem. Rev. 2019, 400, 213042. [Google Scholar] [CrossRef]
  76. Donato, G.; Grosvenor, A.P. Crystallization of rare-earth phosphate-borosilicate glass composites synthesized by a one-step coprecipitation method. Cryst. Growth Des. 2020, 20, 2217–2231. [Google Scholar] [CrossRef]
  77. Aleshin, D.K.; Mashkovtsev, M.A.; Kuznetsova, Y.A.; Rychkov, V.N.; Zatsepin, A.F.; Gordeev, E.V. Fabrication of (Y0.95Eu0.05)2O3 phosphors with enhanced properties by coprecipitation of layered rare earth hydroxide. J. Alloys Compd. 2019, 805, 258–266. [Google Scholar] [CrossRef]
  78. Yi, G.S.; Chow, G.M. Colloidal LaF3: Yb, Er, LaF3: Yb, Ho and LaF3: Yb, Tm nanocrystals with multicolor upconversion fluorescence. J. Mater. Chem. 2005, 15, 4460–4464. [Google Scholar] [CrossRef]
  79. Guan, H.; Feng, Y.; Zhang, W.; Wang, W.; Hu, Y. Room-temperature facile synthesis of hexagonal NaYF4 and NaYF4: Yb, Er powder without any organic additives and its upconversion fluorescence properties. Adv. Powder Technol. 2022, 33, 103381. [Google Scholar] [CrossRef]
  80. Sarkar, D.; Meesaragandla, B.; Samanta, T.; Mahalingam, V. A greener approach towards making highly luminescent Ln3+-doped NaYF4 nanoparticles with ligand-assisted phase control. ChemistrySelect 2016, 1, 4785–4793. [Google Scholar] [CrossRef]
  81. Wang, M.; Mi, C.C.; Wang, S.; Li, F.; Liu, J.L.; Xu, S.K. Synthesis and characterization of NaYF4: Yb, Er upconversion fluorescent nanoparticles via a co-precipitation method. Spectrosc. Spect. Anal. 2009, 29, 3327–3331. [Google Scholar]
  82. Sekiyama, S.; Umezawa, M.; Kuraoka1, S.; Ube, T.; Kamimura, M.; Soga, K. Temperature sensing of deep abdominal region in mice by using over-1000 nm near-infrared luminescence of rare-earth-doped NaYF4 nanothermometer. Sci. Rep. 2018, 8, 16979. [Google Scholar] [CrossRef] [PubMed]
  83. Kong, M.; Gu, Y.Y.; Chai, Y.; Ke, J.; Liu, Y.; Xu, X.; Li, Z.X.; Feng, W.; Li, F.Y. Luminescence interference-free lifetime nanothermometry pinpoints in vivo temperature. Sci. China Chem. 2021, 64, 974–984. [Google Scholar] [CrossRef]
  84. Wang, Z.; Xu, H.; Jia, M.; Jin, X.; Lv, Z.; Dai, M.; Zhu, K.M.; Feng, J.; Ge, X.; Fu, Z. Multifunctional lanthanide ions-doped Ba2TiGe2O8 phosphor for near-infrared ratiometric thermometer and information security. J. Lumin. 2022, 243, 118652. [Google Scholar] [CrossRef]
  85. Nexha, A.; Carvajal, J.J.; Pujol, M.C.; Díaza, F.; Aguiló, M. Lanthanide doped luminescence nanothermometers in the biological windows: Strategies and applications. Nanoscale 2021, 13, 7913–7987. [Google Scholar] [CrossRef]
  86. Lei, X.; Li, R.; Tu, D.; Shang, X.Y.; Liu, Y.; You, W.; Sun, C.; Zhang, F.; Chen, X.Y. Intense near-infrared-Ⅱ luminescence from NaCeF4: Er/Yb nanoprobes for in vitro bioas-say and in vivo bioimaging. Chem. Sci. 2018, 9, 4682–4688. [Google Scholar] [CrossRef] [Green Version]
  87. Wang, C.; Niu, M.; Wang, W.; Su, L.; Feng, H.J.; Lin, H.X.; Ge, X.; Wu, R.; Li, Q.; Liu, J.; et al. In situ activatable ratiometric NIR-II fluorescence nanoprobe for quantitative detection of H2S in colon cancer. Anal. Chem. 2021, 93, 9356–9363. [Google Scholar] [CrossRef]
  88. Jia, Q.; Liu, Y.; Duan, Y.; Zhou, J. Interference-free detection of hydroxyl radical and arthritis diagnosis by rare earth-based nanoprobe utilizing SWIR emission as reference. Anal. Chem. 2019, 91, 11433–11439. [Google Scholar] [CrossRef]
  89. Liao, N.; Su, L.; Zheng, Y.; Zhao, B.; Wu, M.; Zhang, D.; Yang, H.H.; Liu, X.L.; Song, J.B. In vivo tracking of cell viability for adoptive natural killer cell-based immunotherapy by ratiometric NIR-II fluorescence imaging. Angew. Chem. Int. Ed. 2021, 133, 2–11. [Google Scholar] [CrossRef]
  90. Liao, N.; Su, L.; Cao, Y.; Qiu, L.; Xie, R.; Peng, F.; Cai, Z.; Liu, X.; Song, J.; Zeng, Y. Tracking cell viability for adipose-derived mesenchymal stem cell-based therapy by quantitative fluorescence imaging in the second near-infrared window. ACS Nano 2022, 16, 2889–2900. [Google Scholar] [CrossRef]
  91. Wang, S.; Liu, L.; Fan, Y.; El-Toni, A.M.; Alhoshan, M.S.; Li, D.; Zhang, F. In vivo high-resolution ratiometric fluorescence imaging of inflammation using NIR-II nanoprobes with 1550 nm emission. Nano Lett. 2019, 19, 2418–2427. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, M.; Wang, Z.; Wang, C.; Wu, Y.T.; Li, Z.; Liu, Z.H. Visualizing oxidative stress level for timely assessment of ischemic stroke via a ratiometric near-infrared-II luminescent nanoprobe. ACS Nano 2021, 15, 11940–11952. [Google Scholar] [CrossRef] [PubMed]
  93. Cao, C.; Zhou, X.B.; Xue, M.; Han, C.; Feng, W.; Li, F.Y. Dual near-infrared-emissive luminescent nanoprobes for ratiometric luminescent monitoring of ClO in living organisms. ACS Appl. Mater. Interfaces 2019, 11, 15298–15305. [Google Scholar] [CrossRef]
  94. Ge, X.; Lou, Y.; Su, L.; Chen, B.; Guo, Z.Y.; Gao, S.; Zhang, W.; Chen, T.; Song, J.B.; Yang, H.H. Single wavelength laser excitation ratiometric NIR-II fluorescent probe for molecule imaging in vivo. Anal. Chem. 2020, 92, 6111–6120. [Google Scholar] [CrossRef] [PubMed]
  95. Pei, P.; Hu, H.; Chen, Y.; Wang, S.; Chen, J.; Ming, J.; Yang, Y.; Sun, C.; Zhao, S.; Zhang, F. NIR-II ratiometric lanthanide-dye hybrid nanoprobes doped bioscaffolds for in situ bone repair monitoring. Nano Lett. 2022, 22, 783–791. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, C.; Lin, H.; Ge, X.; Mu, J.; Su, L.; Zhang, X.; Niu, M.; Yang, H.H.; Song, J.B. Dye-sensitized downconversion nanoprobes with emission beyond 1500 nm for ratiometric visualization of cancer redox state. Adv. Funct. Mater. 2021, 31, 2009942. [Google Scholar] [CrossRef]
  97. Li, Z.; Wu, J.; Wang, Q.; Liang, T.; Ge, J.; Wang, P.; Liu, Z. A universal strategy to construct lanthanide-doped nanoparticles-based activable NIR-II luminescence probe for bioimaging. iScience 2020, 23, 100962. [Google Scholar] [CrossRef] [Green Version]
  98. Zhao, M.; Li, B.H.; Wu, Y.; He, H.; Zhu, X.; Zhang, H.; Dou, C.; Feng, L.; Fan, Y.; Zhang, F. A tumor-microenvironment-responsive lanthanide-cyanine FRET sensor for NIR-II luminescence-lifetime in situ imaging of hepatocellular carcinoma. Adv. Mater. 2020, 32, 2001172. [Google Scholar] [CrossRef]
  99. Zhong, Y.; Gu, J.; Su, Y.; Zhao, L.; Zhou, Y.; Peng, J. Real-time screening of hepatotoxins in natural medicine by peroxynitrite responsive lanthanide-based NIR-II luminescent probes. Chem. Eng. J. 2022, 433, 133263. [Google Scholar] [CrossRef]
  100. Liu, Q.; Zhong, Y.; Su, Y.; Zhao, L.Z.; Peng, J.J. Real-time imaging of hepatic inflammation using hydrogen sulfide-activatable second near-infrared luminescent nanoprobes. Nano Lett. 2021, 21, 4606–4614. [Google Scholar] [CrossRef]
  101. Deng, Z.; Bi, S.; Jiang, M.Y.; Zeng, S.J. Endogenous H2S-activated orthogonal second near-infrared emissive nanoprobe for in situ ratiometric fluorescence imaging of metformin-induced liver injury. ACS Nano 2021, 15, 3201–3211. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, R.; Zhou, L.; Wang, W.; Li, X.M.; Zhang, F. In vivo gastrointestinal drug-release monitoring through second near-infrared window fluorescent bioimaging with orally delivered microcarriers. Nat. Commun. 2017, 8, 14702. [Google Scholar] [CrossRef] [PubMed]
  103. Su, Y.; Ye, L.; Gu, J.; Zhao, L.; Zhou, Y.; Peng, J. Sensing and imaging of PPi in vivo using lanthanide-based second near-infrared luminescent probes. J. Mater. Chem. B 2022, 10, 1055. [Google Scholar] [CrossRef] [PubMed]
  104. Li, C.Y.; Li, W.F.; Liu, H.H.; Zhang, Y.J.; Chen, G.C.; Li, Z.J.; Wang, Q.B. An activatable NIR-II nanoprobe for in vivo early real-time diagnosis of traumatic brain injury. Angew. Chem. Int. Ed. 2020, 59, 247–252. [Google Scholar] [CrossRef]
  105. Wu, Z.F.; Ke, J.X.; Liu, Y.S.; Sun, P.M.; Hong, M.C. Lanthanide-based NIR-II fluorescent nanoprobes and their biomedical applications. Acta Chim. Sin. 2022, 80, 542–552. [Google Scholar] [CrossRef]
Figure 1. (a) Autofluorescence spectra of mouse liver (black), spleen (red), and heart tissue (blue) under 808 nm light. Inset shows the enlarged results at longer wavelengths. (b) Scattering coefficients of intralipid tissue phantoms and various biological tissues in the wavelength range of 400~1700 nm. (a,b) Adapted, with permission, from Ref. [14]. Copyright 2017, Springer Nature. (c) Absorbance spectra of various biological tissues. Adapted, with permission, from Ref. [15]. Copyright 2013, Springer Nature. (d) Line profile analysis of the fluorescence intensities in the lymph system of a mouse. The values of the S/B express signal to background ratios in the fluorescence intensities. Adapted, with permission, from Ref. [16]. Copyright 2014, Royal Society of Chemistry. (e) Schematic of the imaging depth in tissues for visible, NIR−I, and NIR−II light, respectively.
Figure 1. (a) Autofluorescence spectra of mouse liver (black), spleen (red), and heart tissue (blue) under 808 nm light. Inset shows the enlarged results at longer wavelengths. (b) Scattering coefficients of intralipid tissue phantoms and various biological tissues in the wavelength range of 400~1700 nm. (a,b) Adapted, with permission, from Ref. [14]. Copyright 2017, Springer Nature. (c) Absorbance spectra of various biological tissues. Adapted, with permission, from Ref. [15]. Copyright 2013, Springer Nature. (d) Line profile analysis of the fluorescence intensities in the lymph system of a mouse. The values of the S/B express signal to background ratios in the fluorescence intensities. Adapted, with permission, from Ref. [16]. Copyright 2014, Royal Society of Chemistry. (e) Schematic of the imaging depth in tissues for visible, NIR−I, and NIR−II light, respectively.
Chemosensors 10 00206 g001
Figure 2. The number of published articles on NIR-II in the past ten years, based on Web of Science.
Figure 2. The number of published articles on NIR-II in the past ten years, based on Web of Science.
Chemosensors 10 00206 g002
Figure 4. (a) Schematic illustration, TEM image and emission spectra of the core and core@shell. Adapted, with permission, from Ref. [48]. Copyright 2018, Royal Society of Chemistry. (b) Schematic illustration, NIR−II spectra and intensity of β-NaErF4@NaYF4. Adapted, with permission, from Ref. [49]. Copyright 2020, American Chemical Society. (c) NIR−II luminescence of NaYF4: Yb3+, Er3+ in different shell paths. Adapted, with permission, from Ref. [50]. Copyright 2016, American Chemical Society.
Figure 4. (a) Schematic illustration, TEM image and emission spectra of the core and core@shell. Adapted, with permission, from Ref. [48]. Copyright 2018, Royal Society of Chemistry. (b) Schematic illustration, NIR−II spectra and intensity of β-NaErF4@NaYF4. Adapted, with permission, from Ref. [49]. Copyright 2020, American Chemical Society. (c) NIR−II luminescence of NaYF4: Yb3+, Er3+ in different shell paths. Adapted, with permission, from Ref. [50]. Copyright 2016, American Chemical Society.
Chemosensors 10 00206 g004
Figure 5. (a) Schematic illustrations of the energy transfer pathway from dye on the surface of LnNCs. Adapted, with permission, from Ref. [54]. Copyright 2016, American Chemical Society. (b) NIR−II fluorescence emission of LnNCs with different concentrations of ICG. (c) The luminescence photographs of LnNCs with ICG. (d) The mechanism of ICG-sensitized LnNCs. (bd) Adapted, with permission, from Ref. [55]. Copyright 2018, Wiley−VCH.
Figure 5. (a) Schematic illustrations of the energy transfer pathway from dye on the surface of LnNCs. Adapted, with permission, from Ref. [54]. Copyright 2016, American Chemical Society. (b) NIR−II fluorescence emission of LnNCs with different concentrations of ICG. (c) The luminescence photographs of LnNCs with ICG. (d) The mechanism of ICG-sensitized LnNCs. (bd) Adapted, with permission, from Ref. [55]. Copyright 2018, Wiley−VCH.
Chemosensors 10 00206 g005
Figure 6. (a) Schematic illustration of an inorganic ion of LnNCs sensing. (b) Schematic of organic dyes on the surface of LnNCs for sensing by affecting the luminescence of LnNCs themselves. (c) Schematic of sensing that the surface organic dyes of LnNCs indirectly affect the luminescence by influencing the excitation light source.
Figure 6. (a) Schematic illustration of an inorganic ion of LnNCs sensing. (b) Schematic of organic dyes on the surface of LnNCs for sensing by affecting the luminescence of LnNCs themselves. (c) Schematic of sensing that the surface organic dyes of LnNCs indirectly affect the luminescence by influencing the excitation light source.
Chemosensors 10 00206 g006
Figure 7. (a) Schematic of in sensing with NaCeF4: Er, Yb nanoprobes. (b) NIR−II spectra of NaCeF4: Er, Yb with different H2O2 concentrations. (c) NIR−II spectra of NaCeF4: Er, Yb after the addition of H2O, uricase, UA, and uricase + UA, respectively. Adapted, with permission, from Ref. [86]. Copyright 2018, Royal Society of Chemistry. (d) Schematic diagram of the endogenous H2S−triggered in situ formation of Ag2S in the LnNCs@HSA−Ag+ nanoprobe for the NIR−II. (e,f) NIR−II spectra of the nanoprobe under 808 nm and 980 nm, respectively. Adapted, with permission, from Ref. [87]. Copyright 2021, American Chemical Society.
Figure 7. (a) Schematic of in sensing with NaCeF4: Er, Yb nanoprobes. (b) NIR−II spectra of NaCeF4: Er, Yb with different H2O2 concentrations. (c) NIR−II spectra of NaCeF4: Er, Yb after the addition of H2O, uricase, UA, and uricase + UA, respectively. Adapted, with permission, from Ref. [86]. Copyright 2018, Royal Society of Chemistry. (d) Schematic diagram of the endogenous H2S−triggered in situ formation of Ag2S in the LnNCs@HSA−Ag+ nanoprobe for the NIR−II. (e,f) NIR−II spectra of the nanoprobe under 808 nm and 980 nm, respectively. Adapted, with permission, from Ref. [87]. Copyright 2021, American Chemical Society.
Chemosensors 10 00206 g007
Figure 8. (a,b) Schematic design of the [email protected] for ClO detection. Adapted, with permission, from Ref. [91]. Copyright 2019, American Chemical Society. (c) Schematic illustration of the LnNCs@Cy925 for ClO detection. Adapted, with permission, from Ref. [93]. Copyright 2019, American Chemical Society. (d) Schematic illustration of the LnNCs@NPh for GSH detection. (e,f) NIR−II spectra of the LnNCs@NPh under 808 nm and 980 nm, respectively. Adapted, with permission, from Ref. [96]. Copyright 2021, Wiley−VCH.
Figure 8. (a,b) Schematic design of the [email protected] for ClO detection. Adapted, with permission, from Ref. [91]. Copyright 2019, American Chemical Society. (c) Schematic illustration of the LnNCs@Cy925 for ClO detection. Adapted, with permission, from Ref. [93]. Copyright 2019, American Chemical Society. (d) Schematic illustration of the LnNCs@NPh for GSH detection. (e,f) NIR−II spectra of the LnNCs@NPh under 808 nm and 980 nm, respectively. Adapted, with permission, from Ref. [96]. Copyright 2021, Wiley−VCH.
Chemosensors 10 00206 g008
Figure 9. (a) Schematic design of the LnNCs@MY−1057 for ONOO detection. (b) Luminescence emission intensity of LnNCs@MY−1057. (c) Lifetime response of LnNCs@MY-1057. (d) Plot of intensity changes under different penetration depths. (e) Plot of lifetime changes under different penetration depths. Adapted, with permission, from Ref. [98]. Copyright 2020, Wiley−VCH. (f) Schematic design of the LnNCs@A1094 for ONOO detection. (g) Absorption spectra and (h) NIR−II spectra of LnNCs@A1094 upon gradual addition of ONOO. (i) Plot of fluorescence ratio (I1060/I1525) changes as a function of ONOO concentration. Adapted, with permission, from Ref. [51]. Copyright 2021, American Chemical Society.
Figure 9. (a) Schematic design of the LnNCs@MY−1057 for ONOO detection. (b) Luminescence emission intensity of LnNCs@MY−1057. (c) Lifetime response of LnNCs@MY-1057. (d) Plot of intensity changes under different penetration depths. (e) Plot of lifetime changes under different penetration depths. Adapted, with permission, from Ref. [98]. Copyright 2020, Wiley−VCH. (f) Schematic design of the LnNCs@A1094 for ONOO detection. (g) Absorption spectra and (h) NIR−II spectra of LnNCs@A1094 upon gradual addition of ONOO. (i) Plot of fluorescence ratio (I1060/I1525) changes as a function of ONOO concentration. Adapted, with permission, from Ref. [51]. Copyright 2021, American Chemical Society.
Chemosensors 10 00206 g009
Figure 10. (a) Schematic illustration for the synthesis route of 1−PEI−LnNCs. (b) NIR−II luminescence spectra of 1−PEI−LnNCs. (c) NIR-II spectra of 1-PEI-LnNCs upon gradual addition of Na2S. Adapted, with permission, from Ref. [100]. Copyright 2021, American Chemical Society. (d) Rational design of activatable orthogonal NIR−II emitting LnNCs@SiO2@Ag nanoprobes for H2S. (e) Activatable ratiometric fluorescence probe with H2S responsive turn-on orthogonal NIR−II emission. (f,g) NIR−II spectra by adding different concentrations of Na2S under 980 nm and 808 nm, respectively. Adapted, with permission, from Ref. [101]. Copyright 2021, American Chemical Society.
Figure 10. (a) Schematic illustration for the synthesis route of 1−PEI−LnNCs. (b) NIR−II luminescence spectra of 1−PEI−LnNCs. (c) NIR-II spectra of 1-PEI-LnNCs upon gradual addition of Na2S. Adapted, with permission, from Ref. [100]. Copyright 2021, American Chemical Society. (d) Rational design of activatable orthogonal NIR−II emitting LnNCs@SiO2@Ag nanoprobes for H2S. (e) Activatable ratiometric fluorescence probe with H2S responsive turn-on orthogonal NIR−II emission. (f,g) NIR−II spectra by adding different concentrations of Na2S under 980 nm and 808 nm, respectively. Adapted, with permission, from Ref. [101]. Copyright 2021, American Chemical Society.
Chemosensors 10 00206 g010
Figure 11. (a) Schematic of in vivo mSiO2−Nd tracking and drug release monitoring by NIR−II emission. (b) Experimental processes of designing the NIR−II mesoporous microcarriers. (c) Absorption spectra of LnNCs dispersed in hexane and NPTAT dispersed in water. (d) NIR−II signals, as a function of the NPTAT loading amounts in the microcarrier, under 730 nm excitation. (e) In vivo release percentages of BSA−NPTAT. Adapted, with permission, from Ref. [102]. Copyright 2017, Springer Nature.
Figure 11. (a) Schematic of in vivo mSiO2−Nd tracking and drug release monitoring by NIR−II emission. (b) Experimental processes of designing the NIR−II mesoporous microcarriers. (c) Absorption spectra of LnNCs dispersed in hexane and NPTAT dispersed in water. (d) NIR−II signals, as a function of the NPTAT loading amounts in the microcarrier, under 730 nm excitation. (e) In vivo release percentages of BSA−NPTAT. Adapted, with permission, from Ref. [102]. Copyright 2017, Springer Nature.
Chemosensors 10 00206 g011
Table 1. Typical NIR-II LnNCs-based Bio/chemo sensors.
Table 1. Typical NIR-II LnNCs-based Bio/chemo sensors.
NIR-II LnNCsExcitation (nm)Emission (nm)Response UnitInfluence SignalApplicationsRef.
NaCeF4: Er, Yb9801530Ce3+ → Ce4+1530H2O2[86]
NaErF4: 2%Ho@NaYF41530980/1180IR1061980H2O2[44]
NaErF4@NaLuF4980654/1550Cypate654·OH[88]
NaYF4: 18%Yb3+, 2%Er3+808/9801550IR786s808/1550ROS[89]
NaYF4: 20% Yb and 2%Er@NaYF4808/9801550IR786s808/1550ROS[90]
NaYF4: 50%Er@NaYF4808/9801550Cy7.5808/1550HOCl[91]
NaYbF4: 5%Er, 5%Ce@NaYF4: 20%Nd808/9801550IR-783808/1550HOCl and ·OH[92]
NaYbF4: Er@NaYF4: Yb@NaYF4: Nd808925/1525Cy925925HOCl[93]
NaYF4: 18%Yb3+, 2%Er3+9801150/1550SeTT1150HOCl[94]
NaErF4@NaYF4808/9801525IR808808/1525HOCl[95]
NaYF4: 20%Yb, 2%Er@NaYF4: 30%Nd808/98015504-nitrophenol-Cy7808/1550GSH[96]
NaYF4: 20%Yb, 2%Er@NaYF4: 30%Nd8081530HC-Ni1530GSH[97]
NaYF4@NaYF4: 1%Nd3+8081064MY-10571060ONOO[98]
NaErF4@NaYF4@NaYF4: 10%Nd@NaYF48081060/1525A10941060ONOO[51]
NaGdF4: 3%Nd@NaGdF48081060Compound 11060ONOO[99]
NaYF4: 20%Yb, 2%Er@NaYF4808/9801050/1550Ag+ → Ag2S808/1050H2S[87]
NaGdF4: 2%Nd@NaGdF48081060Compound 11060H2S[100]
NaYF4: Gd, Yb, Er@NaYF4: Yb808/9801053/1525Ag → Ag2S808/1053H2S[101]
NaGdF4: 5%Nd@NaGdF4730/8081064BSA-NPTAT730/1064BSA drug-release monitoring[102]
NaGdF4:Nd@NaGdF48081058TA-Fe3+1058PPi[103]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yang, T.; Qin, J.; Zhang, J.; Guo, L.; Yang, M.; Wu, X.; You, M.; Peng, H. Recent Progresses in NIR-II Luminescent Bio/Chemo Sensors Based on Lanthanide Nanocrystals. Chemosensors 2022, 10, 206. https://0-doi-org.brum.beds.ac.uk/10.3390/chemosensors10060206

AMA Style

Yang T, Qin J, Zhang J, Guo L, Yang M, Wu X, You M, Peng H. Recent Progresses in NIR-II Luminescent Bio/Chemo Sensors Based on Lanthanide Nanocrystals. Chemosensors. 2022; 10(6):206. https://0-doi-org.brum.beds.ac.uk/10.3390/chemosensors10060206

Chicago/Turabian Style

Yang, Tingyu, Jinglei Qin, Jinling Zhang, Lanying Guo, Mu Yang, Xi Wu, Mei You, and Hongshang Peng. 2022. "Recent Progresses in NIR-II Luminescent Bio/Chemo Sensors Based on Lanthanide Nanocrystals" Chemosensors 10, no. 6: 206. https://0-doi-org.brum.beds.ac.uk/10.3390/chemosensors10060206

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop