Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.756
Filter
Add more filters

Publication year range
1.
Cell ; 177(2): 243-255.e15, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30827682

ABSTRACT

Mammals cannot see light over 700 nm in wavelength. This limitation is due to the physical thermodynamic properties of the photon-detecting opsins. However, the detection of naturally invisible near-infrared (NIR) light is a desirable ability. To break this limitation, we developed ocular injectable photoreceptor-binding upconversion nanoparticles (pbUCNPs). These nanoparticles anchored on retinal photoreceptors as miniature NIR light transducers to create NIR light image vision with negligible side effects. Based on single-photoreceptor recordings, electroretinograms, cortical recordings, and visual behavioral tests, we demonstrated that mice with these nanoantennae could not only perceive NIR light, but also see NIR light patterns. Excitingly, the injected mice were also able to differentiate sophisticated NIR shape patterns. Moreover, the NIR light pattern vision was ambient-daylight compatible and existed in parallel with native daylight vision. This new method will provide unmatched opportunities for a wide variety of emerging bio-integrated nanodevice designs and applications. VIDEO ABSTRACT.


Subject(s)
Nanoparticles/therapeutic use , Photoreceptor Cells, Vertebrate/physiology , Vision, Ocular/physiology , Animals , Female , Infrared Rays , Injections/methods , Light , Male , Mammals/physiology , Mice , Mice, Inbred C57BL , Opsins/metabolism , Retina/metabolism , Retina/physiology , Retinal Cone Photoreceptor Cells/physiology , Vision, Ocular/genetics
2.
Nature ; 630(8016): 368-374, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867128

ABSTRACT

Despite its disordered liquid-like structure, glass exhibits solid-like mechanical properties1. The formation of glassy material occurs by vitrification, preventing crystallization and promoting an amorphous structure2. Glass is fundamental in diverse fields of materials science, owing to its unique optical, chemical and mechanical properties as well as durability, versatility and environmental sustainability3. However, engineering a glassy material without compromising its properties is challenging4-6. Here we report the discovery of a supramolecular amorphous glass formed by the spontaneous self-organization of the short aromatic tripeptide YYY initiated by non-covalent cross-linking with structural water7,8. This system uniquely combines often contradictory sets of properties; it is highly rigid yet can undergo complete self-healing at room temperature. Moreover, the supramolecular glass is an extremely strong adhesive yet it is transparent in a wide spectral range from visible to mid-infrared. This exceptional set of characteristics is observed in a simple bioorganic peptide glass composed of natural amino acids, presenting a multi-functional material that could be highly advantageous for various applications in science and engineering.


Subject(s)
Adhesives , Glass , Oligopeptides , Adhesives/chemistry , Glass/chemistry , Temperature , Vitrification , Water/chemistry , Oligopeptides/chemistry , Tyrosine/chemistry , Light , Infrared Rays
3.
Nature ; 604(7905): 287-291, 2022 04.
Article in English | MEDLINE | ID: mdl-35418635

ABSTRACT

Thermophotovoltaics (TPVs) convert predominantly infrared wavelength light to electricity via the photovoltaic effect, and can enable approaches to energy storage1,2 and conversion3-9 that use higher temperature heat sources than the turbines that are ubiquitous in electricity production today. Since the first demonstration of 29% efficient TPVs (Fig. 1a) using an integrated back surface reflector and a tungsten emitter at 2,000 °C (ref. 10), TPV fabrication and performance have improved11,12. However, despite predictions that TPV efficiencies can exceed 50% (refs. 11,13,14), the demonstrated efficiencies are still only as high as 32%, albeit at much lower temperatures below 1,300 °C (refs. 13-15). Here we report the fabrication and measurement of TPV cells with efficiencies of more than 40% and experimentally demonstrate the efficiency of high-bandgap tandem TPV cells. The TPV cells are two-junction devices comprising III-V materials with bandgaps between 1.0 and 1.4 eV that are optimized for emitter temperatures of 1,900-2,400 °C. The cells exploit the concept of band-edge spectral filtering to obtain high efficiency, using highly reflective back surface reflectors to reject unusable sub-bandgap radiation back to the emitter. A 1.4/1.2 eV device reached a maximum efficiency of (41.1 ± 1)% operating at a power density of 2.39 W cm-2 and an emitter temperature of 2,400 °C. A 1.2/1.0 eV device reached a maximum efficiency of (39.3 ± 1)% operating at a power density of 1.8 W cm-2 and an emitter temperature of 2,127 °C. These cells can be integrated into a TPV system for thermal energy grid storage to enable dispatchable renewable energy. This creates a pathway for thermal energy grid storage to reach sufficiently high efficiency and sufficiently low cost to enable decarbonization of the electricity grid.


Subject(s)
Electricity , Hot Temperature , Infrared Rays , Temperature
4.
Genes Dev ; 33(5-6): 333-347, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30796017

ABSTRACT

SUMOylation (small ubiquitin-like modifier) in the DNA double-strand break (DSB) response regulates recruitment, activity, and clearance of repair factors. However, our understanding of a role for deSUMOylation in this process is limited. Here we identify different mechanistic roles for deSUMOylation in homologous recombination (HR) and nonhomologous end joining (NHEJ) through the investigation of the deSUMOylase SENP2. We found that regulated deSUMOylation of MDC1 prevents excessive SUMOylation and its RNF4-VCP mediated clearance from DSBs, thereby promoting NHEJ. In contrast, we show that HR is differentially sensitive to SUMO availability and SENP2 activity is needed to provide SUMO. SENP2 is amplified as part of the chromosome 3q amplification in many cancers. Increased SENP2 expression prolongs MDC1 focus retention and increases NHEJ and radioresistance. Collectively, our data reveal that deSUMOylation differentially primes cells for responding to DSBs and demonstrates the ability of SENP2 to tune DSB repair responses.


Subject(s)
Cysteine Endopeptidases/metabolism , DNA End-Joining Repair/genetics , DNA Repair/genetics , Homologous Recombination/genetics , Sumoylation/genetics , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/radiation effects , Cysteine Endopeptidases/genetics , DNA Breaks, Double-Stranded , HEK293 Cells , HeLa Cells , Humans , Infrared Rays , Nuclear Proteins/metabolism , Radiation Tolerance/genetics , Signal Transduction/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Valosin Containing Protein/metabolism
5.
Nucleic Acids Res ; 52(7): e36, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38407347

ABSTRACT

Cellular RNA is asymmetrically distributed in cells and the regulation of RNA localization is crucial for proper cellular functions. However, limited chemical tools are available to capture dynamic RNA localization in complex biological systems with high spatiotemporal resolution. Here, we developed a new method for RNA proximity labeling activated by near-infrared (NIR) light, which holds the potential for deep penetration. Our method, termed FAP-seq, utilizes a genetically encoded fluorogen activating protein (FAP) that selectively binds to a set of substrates known as malachite green (MG). FAP binding restricts the rotation of MG and rapidly activates its fluorescence in a wash-free manner. By introducing a monoiodo modification to MG, we created a photosensitizer (MG-HI) with the highest singlet oxygen generation ability among various MG derivatives, enabling both protein and RNA proximity labeling in live cells. New insights are provided in the transcriptome analysis with FAP-seq, while a deeper understanding of the symmetry-breaking structural arrangement of FAP-MG-HI was obtained through molecular dynamics simulations. Overall, our wash-free and NIR light-inducible RNA proximity labeling method (FAP-seq) offers a powerful and versatile approach for investigating complex mechanisms underlying RNA-related biological processes.


Subject(s)
Fluorescent Dyes , Infrared Rays , Photosensitizing Agents , RNA , Rosaniline Dyes , Rosaniline Dyes/chemistry , Photosensitizing Agents/chemistry , Humans , Fluorescent Dyes/chemistry , RNA/chemistry , RNA/metabolism , Singlet Oxygen/metabolism , Singlet Oxygen/chemistry , Molecular Dynamics Simulation , HeLa Cells
6.
Nature ; 565(7739): 343-346, 2019 01.
Article in English | MEDLINE | ID: mdl-30651612

ABSTRACT

Recent advances in photoredox catalysis have made it possible to achieve various challenging synthetic transformations, polymerizations and surface modifications1-3. All of these reactions require ultraviolet- or visible-light stimuli; however, the use of visible-light irradiation has intrinsic challenges. For example, the penetration of visible light through most reaction media is very low, leading to problems in large-scale reactions. Moreover, reactants can compete with photocatalysts for the absorption of incident light, limiting the scope of the reactions. These problems can be overcome by the use of near-infrared light, which has a much higher penetration depth through various media, notably biological tissue4. Here we demonstrate various photoredox transformations under infrared radiation by utilizing the photophysical process of triplet fusion upconversion, a mechanism by which two low-energy photons are converted into a higher-energy photon. We show that this is a general strategy applicable to a wide range of photoredox reactions. We tune the upconversion components to adjust the output light, accessing both orange light and blue light from low-energy infrared light, by pairwise manipulation of the sensitizer and annihilator. We further demonstrate that the annihilator itself can be used as a photocatalyst, thus simplifying the reaction. This approach enables catalysis of high-energy transformations through several opaque barriers using low-energy infrared light.


Subject(s)
Infrared Rays , Photochemical Processes/radiation effects , Catalysis/radiation effects , Color , Oxidation-Reduction/radiation effects
7.
Nano Lett ; 24(27): 8287-8295, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38941514

ABSTRACT

Organic dyes with simultaneously boosted near-infrared-II (NIR-II) fluorescence, type I photodynamic therapy (PDT), and photothermal therapy (PTT) in the aggregate state are still elusive due to the unclear structure-function relationship. Herein, electron-withdrawing substituents are introduced at the 5-indolyl positions of BODIPY dyes to form tight J-aggregates for enhanced NIR-II fluorescence and type I PDT/PTT. The introduction of an electron-rich julolidine group at the meso position and an electron-withdrawing substituent (-F) at the indolyl moiety can enhance intermolecular charge transfer and the hydrogen bonding effect, contributing to the efficient generation of superoxide radicals in the aggregate state. The nanoparticles of BDP-F exhibit NIR-II fluorescence at 1000 nm, good superoxide radical generation ability, and a high photothermal conversion efficiency (50.9%), which enabled NIR-II fluorescence-guided vasculature/tumor imaging and additive PDT/PTT. This work provides a strategy for constructing phototheranostic agents with enhanced NIR-II fluorescence and type I PDT/PTT for broad biomedical applications.


Subject(s)
Boron Compounds , Fluorescent Dyes , Photochemotherapy , Boron Compounds/chemistry , Boron Compounds/therapeutic use , Humans , Fluorescent Dyes/chemistry , Animals , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms/diagnostic imaging , Neoplasms/therapy , Cell Line, Tumor , Optical Imaging/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photothermal Therapy , Electrons , Infrared Rays , Fluorescence
8.
Nano Lett ; 24(25): 7764-7773, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38864366

ABSTRACT

Inducing immunogenic cell death (ICD) during photothermal therapy (PTT) has the potential to effectively trigger photothermal immunotherapy (PTI). However, ICD induced by PTT alone is often limited by inefficient PTT, low immunogenicity of tumor cells, and a dysregulated redox microenvironment. Herein, we develop MoSe2 nanosheets with high-percentage metallic 1T phase and rich exposed active Mo centers through phase and defect engineering of MoSe2 as an effective nanoagent for PTI. The metallic 1T phase in MoSe2 nanosheets endows them with strong PTT performance, and the abundant exposed active Mo centers endow them with high activity for glutathione (GSH) depletion. The MoSe2-mediated high-performance PTT synergizing with efficient GSH depletion facilitates the release of tumor-associated antigens to induce robust ICD, thus significantly enhancing checkpoint blockade immunotherapy and activating systemic immune response in mouse models of colorectal cancer and triple-negative metastatic breast cancer.


Subject(s)
Immunotherapy , Molybdenum , Photothermal Therapy , Animals , Mice , Immunotherapy/methods , Humans , Molybdenum/chemistry , Female , Cell Line, Tumor , Nanostructures/chemistry , Nanostructures/therapeutic use , Glutathione/chemistry , Glutathione/metabolism , Colorectal Neoplasms/therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Immunogenic Cell Death/drug effects , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Infrared Rays , Selenium/chemistry , Selenium/therapeutic use , Phototherapy/methods
9.
Neuroimage ; 292: 120615, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38631617

ABSTRACT

Functional near-infrared spectroscopy (fNIRS) studies often aim to measure changes in the brain's hemodynamic response in relation to a specific intervention. We recently showed how a fNIRS device could induce photobiomodulatory effects on cognition by using its near-infrared (NIR) light. However, so far, fNIRS research has overlooked the stimulatory potential intrinsic to this technique. The work by Kuwamizu et al. (2023) on pupil dynamics during exercise is no exception. Here, we suggest a fix to their experimental design, which could be taken into account in other fNIRS studies, to guarantee an adequate level of control for possible unconsidered photobiomodulatory effects.


Subject(s)
Cognition , Exercise , Spectroscopy, Near-Infrared , Spectroscopy, Near-Infrared/methods , Humans , Exercise/physiology , Cognition/physiology , Infrared Rays , Brain/physiology , Brain/diagnostic imaging , Functional Neuroimaging/methods
10.
J Am Chem Soc ; 146(17): 11669-11678, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38644738

ABSTRACT

Accurate in vivo imaging of G-quadruplexes (G4) is critical for understanding the emergence and progression of G4-associated diseases like cancer. However, existing in vivo G4 fluorescent probes primarily operate within the near-infrared region (NIR-I), which limits their application accuracy due to the short emission wavelength. The transition to second near-infrared (NIR-II) fluorescent imaging has been of significant interest, as it offers reduced autofluorescence and deeper tissue penetration, thereby facilitating more accurate in vivo imaging. Nonetheless, the advancement of NIR-II G4 probes has been impeded by the absence of effective probe design strategies. Herein, through a "step-by-step" rational design approach, we have successfully developed NIRG-2, the first small-molecule fluorescent probe with NIR-II emission tailored for in vivo G4 detection. Molecular docking calculations reveal that NIRG-2 forms stable hydrogen bonds and strong π-π interactions with G4 structures, which effectively inhibit twisted intramolecular charge transfer (TICT) and, thereby, selectively illuminate G4 structures. Due to its NIR-II emission (940 nm), large Stokes shift (90 nm), and high selectivity, NIRG-2 offers up to 47-fold fluorescence enhancement and a tissue imaging depth of 5 mm for in vivo G4 detection, significantly outperforming existing G4 probes. Utilizing NIRG-2, we have, for the first time, achieved high-contrast visualization of tumor metastasis through lymph nodes and precise tumor resection. Furthermore, NIRG-2 proves to be highly effective and reliable in evaluating surgical and drug treatment efficacy in cancer lymphatic metastasis models. We are optimistic that this study not only provides a crucial molecular tool for an in-depth understanding of G4-related diseases in vivo but also marks a promising strategy for the development of clinical NIR-II G4-activated probes.


Subject(s)
Fluorescent Dyes , G-Quadruplexes , Optical Imaging , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Humans , Animals , Neoplasm Metastasis , Mice , Molecular Docking Simulation , Drug Design , Infrared Rays , Cell Line, Tumor , Molecular Structure
11.
J Am Chem Soc ; 146(22): 15251-15263, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38780071

ABSTRACT

Glioblastoma (GBM) poses a significant therapeutic challenge due to its invasive nature and limited drug penetration through the blood-brain barrier (BBB). In response, here we present an innovative biomimetic approach involving the development of genetically engineered exosome nanocatalysts (Mn@Bi2Se3@RGE-Exos) for efficient GBM therapy via improving the BBB penetration and enzyme-like catalytic activities. Interestingly, a photothermally activatable multiple enzyme-like reactivity is observed in such a nanosystem. Upon NIR-II light irradiation, Mn@Bi2Se3@RGE-Exos are capable of converting hydrogen peroxide into hydroxyl radicals, oxygen, and superoxide radicals, providing a peroxidase (POD), oxidase (OXD), and catalase (CAT)-like nanocatalytic cascade. This consequently leads to strong oxidative stresses to damage GBM cells. In vitro, in vivo, and proteomic analysis further reveal the potential of Mn@Bi2Se3@RGE-Exos for the disruption of cellular homeostasis, enhancement of immunological response, and the induction of cancer cell ferroptosis, showcasing a great promise in anticancer efficacy against GBM with a favorable biosafety profile. Overall, the success of this study provides a feasible strategy for future design and clinical study of stimuli-responsive nanocatalytic medicine, especially in the context of challenging brain cancers like GBM.


Subject(s)
Exosomes , Glioblastoma , Infrared Rays , Phototherapy , Glioblastoma/drug therapy , Glioblastoma/therapy , Humans , Exosomes/chemistry , Exosomes/metabolism , Animals , Phototherapy/methods , Mice , Catalysis , Cell Line, Tumor , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Manganese/chemistry , Manganese/pharmacology , Blood-Brain Barrier/metabolism
12.
Breast Cancer Res ; 26(1): 80, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773552

ABSTRACT

BACKGROUND: Cancer-related fatigue (CRF) is a pervasive, persistent, and distressing symptom experienced by cancer patients, for which few treatments are available. We investigated the efficacy and safety of infrared laser moxibustion (ILM) for improving fatigue in breast cancer survivors. METHODS: A three-arm, randomized, sham-controlled clinical trial (6-week intervention plus 12-week observational follow-up) was conducted at a tertiary hospital in Shanghai, China. The female breast cancer survivors with moderate to severe fatigue were randomized 2:2:1 to ILM (n = 56) sham ILM (n = 56), and Waitlist control (WLC)(n = 28) groups. Patients in the ILM and sham ILM (SILM) groups received real or sham ILM treatment, 2 sessions per week for 6 weeks, for a total of 12 sessions. The primary outcome was change in the Brief Fatigue Inventory (BFI) score from baseline to week 6 with follow-up until week 18 assessed in the intention-to-treat population. RESULTS: Between June 2018 and July 2021, 273 patients were assessed for eligibility, and 140 patients were finally enrolled and included in the intention-to-treat analysis. Compared with WLC, ILM reduced the average BFI score by 0.9 points (95% CI, 0.3 to 1.6, P = .007) from baseline to week 6, with a difference between the groups of 1.1 points (95% CI, 0.4 to 1.8, P = .002) at week 18. Compared with SILM, ILM treatment resulted in a non-significant reduction in the BFI score (0.4; 95% CI, -0.2 to 0.9, P = .206) from baseline to week 6, while the between-group difference was significant at week 18 (0.7; 95% CI, 0.2 to 1.3, P = .014). No serious adverse events were reported. CONCLUSION: While ILM was found to be safe and to significantly reduce fatigue compared with WLC, its promising efficacy against the sham control needs to be verified in future adequately powered trials. TRIAL REGISTRATION: Clinicaltrials.gov: NCT04144309. Registered 12 June 2018.


Subject(s)
Breast Neoplasms , Cancer Survivors , Fatigue , Moxibustion , Humans , Female , Moxibustion/methods , Moxibustion/adverse effects , Breast Neoplasms/complications , Breast Neoplasms/therapy , Fatigue/etiology , Fatigue/therapy , Middle Aged , Treatment Outcome , Adult , Quality of Life , China/epidemiology , Aged , Infrared Rays/therapeutic use
13.
Rep Prog Phys ; 87(6)2024 May 20.
Article in English | MEDLINE | ID: mdl-38701769

ABSTRACT

Infrared (IR) neuromodulation (INM) is an emerging light-based neuromodulation approach that can reversibly control neuronal and muscular activities through the transient and localized deposition of pulsed IR light without requiring any chemical or genetic pre-treatment of the target cells. Though the efficacy and short-term safety of INM have been widely demonstrated in both peripheral and central nervous systems, the investigations of the detailed cellular and biological processes and the underlying biophysical mechanisms are still ongoing. In this review, we discuss the current research progress in the INM field with a focus on the more recently discovered IR nerve inhibition. Major biophysical mechanisms associated with IR nerve stimulation are summarized. As the INM effects are primarily attributed to the spatiotemporal thermal transients induced by water and tissue absorption of pulsed IR light, temperature monitoring techniques and simulation models adopted in INM studies are discussed. Potential translational applications, current limitations, and challenges of the field are elucidated to provide guidance for future INM research and advancement.


Subject(s)
Infrared Rays , Animals , Humans
14.
Cancer Sci ; 115(7): 2396-2409, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38671582

ABSTRACT

Near-infrared photoimmunotherapy (NIR-PIT) is a new type of cancer therapy that employs antibody-IRDye700DX conjugates (AbPCs) and near-infrared (NIR) light at a wavelength of 689 nm, the excitation wavelength of IR700. Administered intravenously, injected AbPCs bind specifically to cells expressing the target antigen, whereupon NIR light exposure causes rapid, selective killing. This process induces an anticancer T cell response, leading to sustained anticancer host immune response. Programmed cell death ligand-1 (PD-L1) is a major inhibitory immune checkpoint molecule expressed in various cancers. In this study, we first assessed the efficacy of PD-L1-targeted NIR-PIT (αPD-L1-PIT) in immune-competent tumor mouse models. αPD-L1-PIT showed a significant therapeutic effect on the tumor models with high PD-L1 expression. Furthermore, αPD-L1-PIT induced an abscopal effect on distant tumors and long-term immunological memory. In contrast, αPD-L1-PIT was not as effective for tumor models with low PD-L1 expression. To improve the efficacy of PD-L1-targeted NIR-PIT, PEGylated interferon-gamma (IFNγ) was administered with αPD-L1-PIT. The combination therapy improved the treatment efficacy by increasing PD-L1 expression leading to more efficient cell killing by αPD-L1-PIT. Furthermore, the PEGylated IFNγ led to a CD8+ T cell-dominant tumor microenvironment (TME) with an enhanced anticancer T cell response after αPD-L1-PIT. As a result, even so-called cold tumors exhibited complete responses after αPD-L1-PIT. Thus, combination therapy of PEGylated IFNγ and PD-L1-targeted NIR-PIT has the potential to be an important future strategy for cancer immunotherapy.


Subject(s)
B7-H1 Antigen , Immunotherapy , Infrared Rays , Phototherapy , Tumor Microenvironment , Animals , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Mice , Immunotherapy/methods , Cell Line, Tumor , Phototherapy/methods , Humans , Female , Indoles/pharmacology , Indoles/therapeutic use , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice, Inbred C57BL
15.
Br J Cancer ; 130(10): 1647-1658, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38555315

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME) play a critical role in tumor immunosuppression. However, targeted depletion of CAFs is difficult due to their diverse cells of origin and the resulting lack of specific surface markers. Near-infrared photoimmunotherapy (NIR-PIT) is a novel cancer treatment that leads to rapid cell membrane damage. METHODS: In this study, we used anti-mouse fibroblast activation protein (FAP) antibody to target FAP+ CAFs (FAP-targeted NIR-PIT) and investigated whether this therapy could suppress tumor progression and improve tumor immunity. RESULTS: FAP-targeted NIR-PIT induced specific cell death in CAFs without damaging adjacent normal cells. Furthermore, FAP-targeted NIR-PIT treated mice showed significant tumor regression in the CAF-rich tumor model accompanied by an increase in CD8+ tumor infiltrating lymphocytes (TILs). Moreover, treated tumors showed increased levels of IFN-γ, TNF-α, and IL-2 in CD8+ TILs compared with non-treated tumors, suggesting enhanced antitumor immunity. CONCLUSIONS: Cancers with FAP-positive CAFs in their TME grow rapidly and FAP-targeted NIR-PIT not only suppresses their growth but improves tumor immunosuppression. Thus, FAP-targeted NIR-PIT is a potential therapeutic strategy for selectively targeting the TME of CAF+ tumors.


Subject(s)
Cancer-Associated Fibroblasts , Immunotherapy , Tumor Microenvironment , Animals , Humans , Mice , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Endopeptidases , Gelatinases/metabolism , Immunotherapy/methods , Infrared Rays/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Phototherapy/methods , Serine Endopeptidases/metabolism , Tumor Microenvironment/immunology
16.
Anal Chem ; 96(9): 3772-3779, 2024 03 05.
Article in English | MEDLINE | ID: mdl-38372636

ABSTRACT

Development of new near-infrared fluorophores is one of the eternal themes in the field of biosensing and biological imaging. In this work, we constructed a novel fluorophore platform MOR by replacing methylindole of hemicyanine fluorophore (CyR) with benzoxazole to acquire better fluorescence characteristics. Based on the platform, a near infrared (NIR) fluorescent probe MOR-CES2 was synthesized for the specific "off-on" response to carboxylesterase 2 (CES2). The probe exhibited excellent properties including near-infrared emission (735 nm), large Stokes shift (105 nm), high sensitivity (LOD, 0.3 ng/mL), and rapid response (15 min). The successful application of MOR-CES2 in biological imaging of CES2 in mice with thyroid cancer and inflammatory bowel disease demonstrated that the probe could identify cancer cells and tissues and sensitively respond to inflammation. The results proved the potency of MOR-CES2 as an efficient imaging tool to assist in the surgical resection of CES2-related tumors.


Subject(s)
Fluorescent Dyes , Thyroid Neoplasms , Mice , Animals , Optical Imaging/methods , Thyroid Neoplasms/diagnostic imaging , Infrared Rays
17.
Anal Chem ; 96(18): 7240-7247, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38661330

ABSTRACT

In light of deep tissue penetration and ultralow background, near-infrared (NIR) persistent luminescence (PersL) bioprobes have become powerful tools for bioapplications. However, the inhomogeneous signal attenuation may significantly limit its application for precise biosensing owing to tissue absorption and scattering. In this work, a PersL lifetime-based nanoplatform via deep learning was proposed for high-fidelity bioimaging and biosensing in vivo. The persistent luminescence imaging network (PLI-Net), which consisted of a 3D-deep convolutional neural network (3D-CNN) and the PersL imaging system, was logically constructed to accurately extract the lifetime feature from the profile of PersL intensity-based decay images. Significantly, the NIR PersL nanomaterials represented by Zn1+xGa2-2xSnxO4: 0.4 % Cr (ZGSO) were precisely adjusted over their lifetime, enabling the PersL lifetime-based imaging with high-contrast signals. Inspired by the adjustable and reliable PersL lifetime imaging of ZGSO NPs, a proof-of-concept PersL nanoplatform was further developed and showed exceptional analytical performance for hypochlorite detection via a luminescence resonance energy transfer process. Remarkably, on the merits of the dependable and anti-interference PersL lifetimes, this PersL lifetime-based nanoprobe provided highly sensitive and accurate imaging of both endogenous and exogenous hypochlorite. This breakthrough opened up a new way for the development of high-fidelity biosensing in complex matrix systems.


Subject(s)
Biosensing Techniques , Deep Learning , Hypochlorous Acid , Biosensing Techniques/methods , Hypochlorous Acid/analysis , Luminescence , Infrared Rays , Humans , Animals , Nanostructures/chemistry , Luminescent Measurements/methods , Mice
18.
Anal Chem ; 96(19): 7687-7696, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38693877

ABSTRACT

Smart theranostic nanoprobes with the integration of multiple therapeutic modalities are preferred for precise diagnosis and efficient therapy of tumors. However, it remains a big challenge to arrange the imaging and two or more kinds of therapeutic agents without weakening the intended performances. In addition, most existing fluorescence (FL) imaging agents suffer from low spatiotemporal resolution due to the short emission wavelength (<900 nm). Here, novel three-in-one Ag2S quantum dot (QD)-based smart theranostic nanoprobes were proposed for in situ ratiometric NIR-II FL imaging-guided ion/gas combination therapy of tumors. Under the acidic tumor microenvironment, three-in-one Ag2S QDs underwent destructive degradation, generating toxic Ag+ and H2S. Meanwhile, their FL emission at 1270 nm was weakened. Upon introduction of a downconversion nanoparticle (DCNP) as the delivery carrier and NIR-II FL reference signal unit, the formed Ag2S QD-based theranostic nanoprobes could achieve precise diagnosis of tumors through ratiometric NIR-II FL signals. Also, the generated Ag+ and H2S enabled specific ion/gas combination therapy toward tumors. By combining the imaging and therapeutic functions, three-in-one Ag2S QDs may open a simple yet reliable avenue to design theranostic nanoprobes.


Subject(s)
Optical Imaging , Quantum Dots , Silver Compounds , Quantum Dots/chemistry , Silver Compounds/chemistry , Humans , Animals , Mice , Infrared Rays , Theranostic Nanomedicine , Hydrogen Sulfide/analysis , Hydrogen Sulfide/chemistry , Hydrogen-Ion Concentration
19.
Anal Chem ; 96(3): 1019-1028, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38190738

ABSTRACT

Picosecond infrared laser mass spectrometry (PIRL-MS) is shown, through a retrospective patient tissue study, to differentiate medulloblastoma cancers from pilocytic astrocytoma and two molecular subtypes of ependymoma (PF-EPN-A, ST-EPN-RELA) using laser-extracted lipids profiled with PIRL-MS in 10 s of sampling and analysis time. The average sensitivity and specificity values for this classification, taking genomic profiling data as standard, were 96.41 and 99.54%, and this classification used many molecular features resolvable in 10 s PIRL-MS spectra. Data analysis and liquid chromatography coupled with tandem high-resolution mass spectrometry (LC-MS/MS) further allowed us to reduce the molecular feature list to only 18 metabolic lipid markers most strongly involved in this classification. The identified 'metabolite array' was comprised of a variety of phosphatidic and fatty acids, ceramides, and phosphatidylcholine/ethanolamine and could mediate the above-mentioned classification with average sensitivity and specificity values of 94.39 and 98.78%, respectively, at a 95% confidence in prediction probability threshold. Therefore, a rapid and accurate pathology classification of select pediatric brain cancer types from 10 s PIRL-MS analysis using known metabolic biomarkers can now be available to the neurosurgeon. Based on retrospective mining of 'survival' versus 'extent-of-resection' data, we further identified pediatric cancer types that may benefit from actionable 10 s PIRL-MS pathology feedback. In such cases, aggressiveness of the surgical resection can be optimized in a manner that is expected to benefit the patient's overall or progression-free survival. PIRL-MS is a promising tool to drive such personalized decision-making in the operating theater.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Humans , Child , Chromatography, Liquid , Lipidomics , Retrospective Studies , Infrared Rays , Tandem Mass Spectrometry , Lasers , Brain Neoplasms/diagnosis
20.
Anal Chem ; 96(29): 12065-12073, 2024 07 23.
Article in English | MEDLINE | ID: mdl-38982573

ABSTRACT

Inflammatory bowel disease (IBD) is an idiopathic intestinal inflammatory disease, whose etiology is intimately related to the overproduction of hypochlorous acid (HClO). Optical monitoring of HClO in the living body favors real-time diagnosis of inflammatory diseases. However, HClO-activated near-infrared (NIR) fluorescent probes with rapid response and high inflammatory cell uptake are still lacking. Herein, we report an activatable acceptor-π-acceptor (A-π-A)-type NIR fluorescent probe (Cy-DM) bearing two d-mannosamine groups for the sensitive detection of HClO in early IBD and stool testing. Once reacted with HClO, nonfluorescent Cy-DM could be turned on within 2 s by generating a donor-π-acceptor (D-π-A) structure due to the enhanced intramolecular charge transfer mechanism, showing intense NIR fluorescence emission at 700 nm and a large Stokes shift of 115 nm. Moreover, it was able to sensitively and selectively image exogenous and endogenous HClO in the lysosomes of living cells with a detection limit of 0.84 µM. More importantly, because of the d-mannosamine modification, Cy-DM was efficiently taken up by inflammatory cells in the intestine after intravenous administration, allowing noninvasive visualization of endogenous HClO in a lipopolysaccharide-induced IBD mouse model with a high fluorescence contrast of 6.8/1. In addition, water-soluble Cy-DM has also been successfully applied in ex vivo optical fecal analysis, exhibiting a 3.4-fold higher fluorescence intensity in the feces excreted by IBD mice. We believe that Cy-DM is promising as an invaluable tool for rapid diagnosis of HClO-related diseases as well as stool testing.


Subject(s)
Feces , Fluorescent Dyes , Hypochlorous Acid , Inflammatory Bowel Diseases , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Hypochlorous Acid/analysis , Hypochlorous Acid/metabolism , Inflammatory Bowel Diseases/diagnostic imaging , Animals , Feces/chemistry , Mice , Humans , Optical Imaging , Infrared Rays
SELECTION OF CITATIONS
SEARCH DETAIL