Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Molecules ; 28(4)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36838886

RESUMEN

Accurate detection of H2S is crucial to understanding the occurrence and development of H2S-related diseases. However, the accurate and sensitive detection of H2S in vivo still faces great challenges due to the characteristics of H2S diffusion and short half-life. Herein, we report a H2S-activatable ratiometric near-infrared (NIR) fluorescence liposome nanoprobe HS-CG by the thin-film hydration method. HS-CG shows "always on" fluorescence signal at 816 nm and low fluorescence signal at 728 nm; the NIR fluorescence ratio between 728 and 816 nm (F728/F816) is low. Upon reaction with H2S, the fluorescence at 728 nm could be more rapidly turned on due to strong electrostatic interaction between enriched HS- and positively charged 1,2-dihexadecanoyl-sn-glycero-3-phosphocholine (DPPC) doped in the liposome nanoprobe HS-CG, resulting in a large enhancement of F728/F816, which allows for sensitive visualization of the tumor H2S levels in vivo. This study demonstrates that this strategy of electrostatic adsorption between HS- and positively charged molecules provides a new way to enhance the reaction rate of the probe and H2S, thus serving as an effective platform for improving the sensitivity of imaging.


Asunto(s)
Sulfuro de Hidrógeno , Liposomas , Humanos , Fluorescencia , Colorantes Fluorescentes , Difusión
2.
Angew Chem Int Ed Engl ; 62(10): e202217055, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36602292

RESUMEN

Tumor-targeted and stimuli-activatable nanosensitizers are highly desirable for cancer theranostics. However, designing smart nanosensitizers with multiple imaging signals and synergistic therapeutic activities switched on is challenging. Herein, we report tumor-targeted and redox-activatable nanosensitizers (1-NPs) for sono-photodynamic immunotherapy of tumors by molecular co-assembly and redox-controlled disassembly. 1-NPs show a high longitudinal relaxivity (r1 =18.7±0.3 mM-1 s-1 ), but "off" dual fluorescence (FL) emission (at 547 and 672 nm), "off" sono-photodynamic therapy and indoleamine 2,3-dioxygenase 1 (IDO1) inhibition activities. Upon reduction by glutathione (GSH), 1-NPs rapidly disassemble and remotely release small molecules 2-Gd, Zn-PPA-SH and NLG919, concurrently switching on (1) dual FL emission, (2) sono-photodynamic therapy and (3) IDO1 inhibition activities. After systemic injection, 1-NPs are effective for bimodal FL and magnetic resonance (MR) imaging-guided sono-photodynamic immunotherapy of orthotropic breast and brain tumors in mice under combined ultrasound (US) and 671-nm laser irradiation.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Animales , Ratones , Fotoquimioterapia/métodos , Neoplasias/tratamiento farmacológico , Fluorescencia , Oxidación-Reducción , Inmunoterapia , Línea Celular Tumoral , Fármacos Fotosensibilizantes/uso terapéutico
3.
Analyst ; 146(6): 1865-1871, 2021 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-33480367

RESUMEN

γ-Glutamyltranspeptidase (GGT) is an important aminopeptidase overexpressed in many malignant tumors, and accurate detection of its activity is useful for the diagnosis and treatment of tumors. Herein, we report a GGT-activatable ratiometric fluorescent probe (1) constructed by covalently linking an 'always-on' BODIPY fluorophore with a GGT-activatable near-infrared (NIR) fluorescent substrate. Upon interaction with GGT, the NIR fluorescence at 735 nm in probe 1 is significantly enhanced, while the fluorescence of BODIPY at 517 nm remains unchanged. Using BODIPY fluorescence as an internal standard, significantly enhanced ratiometric fluorescence between 735 nm and 517 nm could be achieved, allowing accurate detection of the activity of GGT in living subjects independent of probe concentration. We demonstrate that probe 1 is feasible for the evaluation of GGT levels in different tumor cells and differentiation of GGT-positive tumor cells from GGT-negative normal tissue cells. Moreover, probe 1 is further applied for the visualization of tumor via noninvasive ratiometric fluorescence imaging of GGT activity, which could facilitate the detection of GGT-positive tumor tissues and study of GGT-related pathological processes.


Asunto(s)
Neoplasias , gamma-Glutamiltransferasa , Colorantes Fluorescentes , Humanos , Neoplasias/diagnóstico por imagen , Imagen Óptica
4.
Angew Chem Int Ed Engl ; 59(46): 20636-20644, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-32686894

RESUMEN

Stimuli-responsive smart photosensitizer (PS) nanoassemblies that allow enhanced delivery and controlled release of PSs are promising for imaging-guided photodynamic therapy (PDT) of tumors. However, the lack of high-sensitivity and spatial-resolution signals and fast washout of released PSs from tumor tissues have impeded PDT efficacy in vivo. Herein, we report tumor targeting, redox-responsive magnetic and fluorogenic PS nanoassemblies (NP-RGD) synthesized via self-assembly of a cRGD- and disulfide-containing fluorogenic and paramagnetic small molecule (1-RGD) for fluorescence/magnetic resonance bimodal imaging-guided tumor PDT. NP-RGD show high r1 relaxivity but quenched fluorescence and PDT activity; disulfide reduction by glutathione (GSH) promotes efficient disassembly into a small-molecule probe (2-RGD) and an organic PS (PPa-SH), which could further bind with intracellular albumin, allowing prolonged retention and cascade activation of fluorescence and PDT to ablate tumors.


Asunto(s)
Magnetismo , Nanopartículas/química , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Animales , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Oxidación-Reducción , Fármacos Fotosensibilizantes/uso terapéutico
5.
Anal Chem ; 91(21): 13639-13646, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31560193

RESUMEN

Activatable chemiluminescent probes that show enhanced chemiluminescence upon interaction with a molecular target of interest have offered promising tools for sensing and bioimaging in terms of low background, high sensitivity, and improved penetration depth in biological tissues. Here, we reported a γ-glutamyl transpeptidase (GGT) activatable chemiluminescent probe for real-time detection of GGT activity in vitro and in living mice. The probe was designed by caging an electron-withdrawing acrylic group-substituted Schaap's phenoxy-dioxetane with a GGT-recognitive substrate (γ-Glu) and a self-immolative linker (p-aminobenzyl alcohol), which was initially chemiluminescence off. Upon interaction with GGT, strong chemiluminescence with a more than 800-fold turn-on ratio could be achieved in aqueous solution, allowing to specifically detect GGT activity with ultrahigh signal-to-background ratio and sensitivity in vitro and in live cells. We demonstrated that the probe was reliable to quantify the GGT in serum, permitting to accurately report the elevated levels of GGT in lipopolysaccharide-treated mouse serum. Moreover, through real-time chemiluminescence imaging of GGT activity, the designed probe was feasible to detect GGT-positive tumors in living mice after intravenous systemic administration. This study demonstrates the high potential of GGT-activatable chemiluminescent probe for serum assays and molecular imaging, which might find wide applications in diagnosis of GGT-related diseases.


Asunto(s)
Luminiscencia , Sondas Moleculares/química , gamma-Glutamiltransferasa/sangre , Animales , Línea Celular , Humanos , Límite de Detección , Ratones , Especificidad por Sustrato
6.
Chembiochem ; 20(4): 474-487, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30062708

RESUMEN

γ-Glutamyltranspeptidase (GGT) is a cell-membrane-bound protease that participates in cellular glutathione and cysteine homeostasis, which are closely related to many physiological and pathological processes. The accurate measurement of GGT activity is useful for the early diagnosis of diseases. In the past few years, many efforts have been made to build optical imaging probes for the detection of GGT activity both in vitro and in vivo. In this Minireview, recent advances in the development of various optical imaging probes for GGT, including activatable fluorescence probes, ratiometric fluorescence probes, and activatable bioluminescence probes, are summarized. This review starts from the instruction of the GGT enzyme and its biological functions, followed by a discussion of activatable fluorescence probes that show off-on fluorescence in response to GGT. GGT-activatable two-photon fluorescence imaging probes with improved imaging depth and spatial resolution are also discussed. Ratiometric fluorescence probes capable of accurately reporting on GGT levels through a self-calibration mechanism are discussed, followed by describing GGT-activatable bioluminescence probes that can offer a high signal-to-background ratio to detect GGT in living mice. Finally, current challenges and further perspectives for the development of molecular imaging probes for GGT are addressed.


Asunto(s)
Colorantes Fluorescentes/química , Imagen Óptica/métodos , gamma-Glutamiltransferasa/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico por imagen , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Luciferina de Luciérnaga/química , Humanos , Microscopía Fluorescente , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , gamma-Glutamiltransferasa/química
7.
Molecules ; 24(17)2019 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-31461829

RESUMEN

In this work, a novel 7-hydroxybenzoxazinone-based fluorescent probe (PBD) for the selective sensing of biothiols is reported. Upon treatment with biothiols, PBD shows a strong fluorescence enhancement (up to 70-fold) and a large Stokes shift (155 nm). Meanwhile, this probe exhibits high resistance to interference from other amino acids and competing species. PBD features good linearity ranges with a low detection limit of 14.5 nM for glutathione (GSH), 17.5 nM for cysteine (Cys), and 80.0 nM for homocysteine (Hcy), respectively. Finally, the potential utility of this probe for biothiol sensing in living HeLa cells is demonstrated.


Asunto(s)
Benzoxazinas/química , Técnicas Biosensibles/métodos , Colorantes Fluorescentes/química , Cisteína/análisis , Glutatión/análisis , Células HeLa , Homocisteína/análisis , Humanos , Límite de Detección , Imagen Óptica
8.
Chemistry ; 24(22): 5707-5722, 2018 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-29068109

RESUMEN

Biocompatible reactions have emerged as versatile tools to build various molecular imaging probes that hold great promise for the detection of biological processes in vitro and/or in vivo. In this Minireview, we describe the recent advances in the development of a firefly luciferin-inspired biocompatible reaction between cyanobenzothiazole (CBT) and cysteine (Cys), and highlight its versatility to label proteins and build multimodality molecular imaging probes. The review starts from the general introduction of biocompatible reactions, which is followed by briefly describing the development of the firefly luciferin-inspired biocompatible chemistry. We then discuss its applications for the specific protein labeling and for the development of multimodality imaging probes (fluorescence, bioluminescence, MRI, PET, photoacoustic, etc.) that enable high sensitivity and spatial resolution imaging of redox environment, furin and caspase-3/7 activity in living cells and mice. Finally, we offer the conclusions and our perspective on the various and potential applications of this reaction. We hope that this review will contribute to the research of biocompatible reactions for their versatile applications in protein labeling and molecular imaging.


Asunto(s)
Caspasas/metabolismo , Ciclobutanos/farmacología , Luciferina de Luciérnaga/química , Proteínas/química , Animales , Benzotiazoles/química , Cisteína/química , Luciferasas de Luciérnaga/metabolismo , Ratones , Imagen Molecular/métodos , Nitrilos/química
9.
Chemistry ; 24(39): 9812-9819, 2018 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-29766578

RESUMEN

Selective imaging and inducing mitochondrial dysfunction in tumor cells using mitochondria-targeting probes has become as a promising approach for cancer diagnosis and therapy. Here, we report the design of a fluorescent berberine analog, dehydroberberine (DH-BBR), as a new mitochondria-targeting probe capable of self-assembling into monodisperse organic nanoparticles (DTNPs) upon integration with a lipophilic counter anion, allowing for enhanced fluorescence imaging and treatment of tumors in living mice. X-ray crystallography revealed that the self-assembly process was attributed to a synergy of different molecular interactions, including π-π stacking, O⋅⋅⋅π interaction and electrostatic interaction between DH-BBR and counter anions. We demonstrated that DTNPs could efficiently enter tumor tissue following intravenous injection and enhance mitochondrial delivery of DH-BBR via an electrostatic interaction driven anion exchange process. Selective accumulation in the mitochondria capable of emitting strong fluorescence and causing mitochondrial dysfunction was achieved, enabling efficient inhibition of tumor growth in living mice. This study demonstrates promise for applying lipophilic anions to control molecular self-assembly and tune antitumor activity of mitochondria-targeting probes, which can facilitate to improve cancer treatment in vivo.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Berberina/química , Colorantes Fluorescentes/química , Mitocondrias/efectos de los fármacos , Animales , Ratones , Mitocondrias/química
10.
Chemistry ; 23(59): 14778-14785, 2017 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-28653778

RESUMEN

γ-Glutamyl transpeptidase (GGT) is a cell-membrane-bound enzyme that is involved in various physiological and pathological processes and is regarded as a potential biomarker for many malignant tumors, precise detection of which is useful for early cancer diagnosis. Herein, a new GGT-activatable near-infrared (NIR) fluorescence imaging probe (GANP) by linking of a GGT-recognitive substrate γ-glutamate (γ-Glu) and a NIR merocyanine fluorophore (mCy-Cl) with a self-immolative linker p-aminobenzyl alcohol (PABA) is reported. GANP was stable under physiological conditions, but could be efficiently activated by GGT to generate ≈100-fold enhanced fluorescence, enabling high sensitivity (detection limit of ≈3.6 mU L-1 ) and specificity for the real-time imaging of GGT activity as well as rapid evaluation of the inhibition efficacy of GGT inhibitors in living tumor cells. Notably, the deep tissue penetration ability of NIR fluorescence could further allow GANP to image GGT in frozen tumor tissue slices with large penetration depth (>100 µm) and in xenograft tumors in living mice. This GGT activatable NIR fluorescence imaging probe could facilitate the study and diagnosis of other GGT-correlated diseases in vivo.


Asunto(s)
Colorantes Fluorescentes/metabolismo , Neoplasias/patología , gamma-Glutamiltransferasa/metabolismo , Animales , Benzopiranos/química , Alcoholes Bencílicos/química , Línea Celular Tumoral , Femenino , Colorantes Fluorescentes/química , Células HCT116 , Humanos , Indoles/química , Ratones , Ratones Desnudos , Microscopía Fluorescente , Neoplasias/diagnóstico por imagen , Espectroscopía Infrarroja Corta , Trasplante Heterólogo , gamma-Glutamiltransferasa/antagonistas & inhibidores
11.
Colloids Surf B Biointerfaces ; 222: 113130, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36623376

RESUMEN

Dendrimers have numerous applications in imaging and drug delivery. Designing a dendrimer diagnostic platform with a well-defined structure and controlled drug delivery is a formidable challenge. Here, we design dendritic polymer-platinum conjugates (G5-PEG-Pt) as pH-responsive nanovesicles for imaging-guided platinum drug delivery. The G5-PEG-Pt have a well-defined structure, intrinsically bright fluorescence, and acid-responsive drug release. The pH-responsive G5-PEG-Pt could rapidly release the platinum drug at acidic pH (5.0) than neutral pH (7.4). The G5-PEG-Pt could enter SKOV-3 human ovarian cancer cells by the endocytosis pathway and exhibited comparative cytotoxicity to free cisplatin. By virtue of the prolonged blood circulation time and the enhanced permeability and retention (EPR) effect, a 4.4-fold higher tumor platinum uptake than that of free cisplatin was achieved, potentially enhancing the therapeutic indexes of the platinum drug. Therefore, these pH-responsive platinum and fluorescent dendrimer conjugates are expected to be potent in vivo cancer optical imaging and therapy platforms.


Asunto(s)
Antineoplásicos , Dendrímeros , Neoplasias Ováricas , Femenino , Humanos , Dendrímeros/química , Cisplatino/farmacología , Polilisina , Doxorrubicina/farmacología , Doxorrubicina/química , Platino (Metal) , Antineoplásicos/química , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/tratamiento farmacológico , Línea Celular Tumoral
12.
ACS Nano ; 17(17): 17468-17475, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37602689

RESUMEN

Biological channels can rapidly and continuously modulate ion transport behaviors in response to external stimuli, which play essential roles in manipulating physiological and pathological processes in cells. Here, to mimic the biological channels, a bionic nanochannel is developed by synergizing a cationic silicon-substituted rhodamine (SiRh) with a glass nanopipette for transmembrane single-cell quantification. Taking the fast and reversible nucleophilic addition reaction between glutathione (GSH) and SiRh, the bionic nanochannel shows a fast and reversible response to GSH, with its inner-surface charges changing between positive and negative charges, leading to a distinct and reversible switch in ionic current rectification (ICR). With the bionic nanochannel, spatiotemporal-resolved operation is performed to quantify endogenous GSH in a single cell, allowing for monitoring of intracellular GSH fluctuation in tumor cells upon photodynamic therapy and ferroptosis. Our results demonstrate that it is a feasible tool for in situ quantification of the endogenous GSH in single cells, which may be adapted to addressing other endogenous biomolecules in single cells by usage of other stimuli-responsive probes.


Asunto(s)
Biónica , Ferroptosis , Vidrio , Glutatión , Transporte Iónico , Rodaminas
13.
Nat Commun ; 14(1): 800, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36781887

RESUMEN

Temporal control of delivery and release of drugs in tumors are important in improving therapeutic outcomes to patients. Here, we report a sequential stimuli-triggered in situ self-assembly and disassembly strategy to direct delivery and release of theranostic drugs in vivo. Using cisplatin as a model anticancer drug, we design a stimuli-responsive small-molecule cisplatin prodrug (P-CyPt), which undergoes extracellular alkaline phosphatase-triggered in situ self-assembly and succeeding intracellular glutathione-triggered disassembly process, allowing to enhance accumulation and elicit burst release of cisplatin in tumor cells. Compared with cisplatin, P-CyPt greatly improves antitumor efficacy while mitigates off-target toxicity in mice with subcutaneous HeLa tumors and orthotopic HepG2 liver tumors after systemic administration. Moreover, P-CyPt also produces activated near-infrared fluorescence (at 710 nm) and dual photoacoustic imaging signals (at 700 and 750 nm), permitting high sensitivity and spatial-resolution delineation of tumor foci and real-time monitoring of drug delivery and release in vivo. This strategy leverages the advantages offered by in situ self-assembly with those of intracellular disassembly, which may act as a general platform for the design of prodrugs capable of improving drug delivery for cancer theranostics.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Profármacos , Animales , Ratones , Cisplatino/farmacología , Medicina de Precisión , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico
14.
J Colloid Interface Sci ; 639: 302-313, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36805755

RESUMEN

The combination of imaging and different therapeutic strategies into one single nanoplatform often demonstrates improved efficacy over monotherapy in cancer treatments. Herein, a multifunctional nanoplatform (labelled as MPRD) based on molybdenum disulfide quantum dots (MoS2 QDs) is developed to achieve enhanced antitumor efficiency by integrating fluorescence imaging, tumor-targeting and synergistic chemo/photodynamic therapy (PDT) into one system. First, polyethylene glycol (PEG)ylated MoS2 QDs (MP) with desirable stability are synthesized via a hydrothermal process using MoS2 QDs and carboxyamino-terminated oligomeric PEG as raw materials. Then, MP were conjugated with arginine-glycine-aspartic acid (RGD) peptide via amidation to form a novel nanocarrier (MPR), which possesses strong blue fluorescence, good biocompatibility and ανß3 receptor-mediated targeting ability. More importantly, MPR generated reactive oxygen species under 808 nm laser activation to realize targeted antitumor PDT. Further doxorubicin (DOX) was loaded onto MPR, which endows MPRD with localized chemotherapy and pH-responsive drug release. The MPRD exhibits improved chemotherapy performance on HepG2 cells (overexpressing integrin ανß3) owing to enhanced cellular uptake mediated by ανß3 receptor and effective drug release triggered by intracellular pH. Notably, MPRD with efficient tumor targeting ability and high chemo/PDT efficacy under NIR laser irradiation is capable of inhibiting HepG2 tumor cell growth both in vitro and in vivo, which is significantly superior to each individual therapy. These findings demonstrate that MPRD holds great potential in effective cancer therapy.


Asunto(s)
Nanopartículas , Nanosferas , Neoplasias , Fotoquimioterapia , Humanos , Molibdeno , Doxorrubicina/farmacología , Imagen Óptica , Concentración de Iones de Hidrógeno , Línea Celular Tumoral , Liberación de Fármacos
15.
Theranostics ; 12(7): 3178-3195, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35547754

RESUMEN

Near-infrared (NIR) fluorescent imaging of both soluble and insoluble Aß species in the brain of Alzheimer's disease (AD) is crucial for the early diagnosis and intervention of AD. To date, a variety of NIR fluorescent probes have been reported for the detection of Aß species. Among these probes, CRANAD-58 was reported to have the capability to detect both soluble and insoluble Aß species, which is vital to monitor the changes of Aß species during the pathological course of the disease. Though CRANAD-58 has shown promise to noninvasively detect Aß species in transgenic AD mice, the emission wavelength (~670 nm) is still too short for further applications. Therefore, new probes with longer emission wavelength and improved physiological properties are in highly demand. Herein, we report the design and engineering of nine donor-acceptor-donor molecules as "off-on" near-infrared fluorescent probes for in vivo imaging of both soluble and insoluble Aß species in living AD mice owing to its improved in vitro properties and in vivo performance. Methods: We report a two-round strategy to develop nine "off-on" NIR fluorescence probes via structural modification of a curcumin analogue-based donor-acceptor-donor architecture. In round one, probes 1 and 2 were synthesized, and probe 2 was identified to be an optimum probe as it showed distinct "off-on" NIR fluorescence at > 690 nm upon binding to Aß monomers, oligomers and aggregates. To further improve the in vivo performance, further structural modification of probe 2 into probes 3-9 was then conducted. The fluorescence response with Aß species and histological staining in vitro and in vivo imaging of Aß species in APP/PS1 transgenic AD mice and age-matched wild-type mice were performed. Results: We demonstrate that, compared to probe 2, probe 9 with improved physiological properties hold the fastest kinetics (~10 min) to produce not only higher brain fluorescence intensity in 10-month-old APP/PS1 transgenic AD mice, but also afford a higher discrepancy in brain fluorescence to discriminate AD mice from wild-type (WT) mice. Probe 9 also hold the ability to detect soluble Aß species in 6-month-old APP/PS1 transgenic mice. Probe 9 was further applied for dynamic visualization of Aß plaques in a skull-thinning 14-month-old APP/PS1 mouse, which revealed its immediate penetration into brain parenchyma and selective labeling of both parenchymal and angiopathic Aß plaques. In addition, probe 9 possessed significantly high attenuation effect on the aggregation of Aß monomers. Conclusion: Our results demonstrate the good potential of probe 9 for longitudinal NIR fluorescence imaging of soluble and insoluble Aß species in APP/PS1 transgenic AD mice, which may act as a useful tool for early diagnosis and intervention of AD.


Asunto(s)
Enfermedad de Alzheimer , Curcumina , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/patología , Curcumina/química , Modelos Animales de Enfermedad , Colorantes Fluorescentes/química , Ratones , Ratones Transgénicos , Placa Amiloide/metabolismo
16.
ACS Nano ; 16(12): 20607-20621, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36508254

RESUMEN

Controlling delivery and release of therapeutic agents to accomplish on-demand synergistic therapy of orthotopic gliomas is desired but challenging. Here, we report a glioma targeting and redox activatable theranostic nanoprobe (Co-NP-RGD1/1) for magnetic resonance (MR) and fluorescence (FL) bimodal imaging-guided on-demand synergistic chemotherapy/photodynamic therapy (Chemo-PDT) of orthotopic gliomas. Co-NP-RGD1/1 is formed via molecular coassembly of two paramagnetic and fluorogenic small-molecule probes CPT-RGD and PPa-RGD at an optimized molar ratio of 1/1, which shows a high longitudinal relaxivity (r1 = 17.0 ± 0.6 mM-1 s-1, 0.5 T) but weak FL emissions and low Chemo-PDT activity. Upon reduction by endogenous glutathione (GSH), Co-NP-RGD1/1 disassemble and release small molecules 2-RGD, chemodrug camptothecin (CPT), and near-infrared (NIR) photosensitizer (PS) PPa-SH that further binds to endogenous albumin to form PPa-SH-albumin complex, allowing to turn on FL, chemotherapeutic efficacy, and PDT activity for synergistic Chemo-PDT of orthotopic U87MG or U251 gliomas in living mice. Moreover, Co-NP-RGD1/1 can also allow noninvasive detection and monitoring of orthotopic brain tumor growth via FL and MR imaging. Findings suggest the potential of cascade coassembly and stimuli-controlled intracellular disassembly strategy for constructing targeted and activatable nanoagents for improving combinational cancer theranostics.


Asunto(s)
Glioma , Nanopartículas , Fotoquimioterapia , Profármacos , Ratones , Animales , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Profármacos/farmacología , Medicina de Precisión , Nanopartículas/uso terapéutico , Glioma/diagnóstico por imagen , Glioma/tratamiento farmacológico , Albúminas , Oligopéptidos , Línea Celular Tumoral
17.
ACS Appl Bio Mater ; 5(2): 747-760, 2022 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-35040617

RESUMEN

Benefiting from its strong cytotoxic features, singlet oxygen (1O2) has garnered considerable research attention in photodynamic therapy (PDT) and thus, plenty of inorganic PDT agents have been recently developed. However, inorganic PDT agents consisting of metal/semiconductor hybrids are surprisingly rare, bearing very low 1O2 quantum yield, and their in vivo PDT applications remain elusive. Herein, we provide an unprecedented report that the Au/MoS2 hybrid under plasmon resonant excitation can sensitize 1O2 generation with a quantum yield of about 0.22, which is much higher than that of the reported hybrid-based photosensitizers (PSs). This significant enhancement in 1O2 quantum yield is attributed to the hot-electron injection from plasmonic AuNPs to MoS2 NSs due to the matched energy levels. Electron paramagnetic resonance (EPR) spectroscopy with spin trapping and spin labeling verifies the plasmonic generation of hot charge carriers and reactive oxygen species such as superoxide and 1O2. This plasmonic PDT agent shows a remarkable photodynamic bacterial inactivation in vitro and anti-cancer therapeutic ability both in vitro and in vivo, which is solely attributed to high 1O2 generation rather than the plasmonic photothermal effect. Hence, plasmonic Au/MoS2 with enhanced 1O2 quantum yield and appreciable in vivo cancer plasmonic PDT performance holds great promise as an inorganic PS to treat near-surface tumors. As a first demonstration of how metal localized surface plasmon resonance could enhance 1O2 generation, the present study opens up promising opportunities for enhancing 1O2 quantum yield of hybrid-based PSs, leading to achieving a high therapeutic index in plasmon PDT.


Asunto(s)
Nanopartículas del Metal , Neoplasias , Fotoquimioterapia , Oro/farmacología , Humanos , Molibdeno , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Oxígeno Singlete/química
18.
ACS Appl Bio Mater ; 4(3): 2033-2043, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35014329

RESUMEN

Mitochondria-targeting probes that allow us to induce and report mitochondrial dysfunction have become promising theranostic agents for cancer; however, the lack of selectivity toward tumor cells over normal tissue cells has impeded the treatment outcome. Herein, we develop 10 fluorescent dehydroberberine derivatives (B1-B10) capable of lighting up mitochondria and exerting moderate cytotoxicity against tumor cells. To enable the selectivity toward tumor cells over normal tissue cells, we introduced a lipophilic anion tetraphenylborate (TPB-) into the most potent compound B3+Cl- to drive molecular self-assembly into monodisperse organic nanoassemblies (B3NPs) in aqueous solution, which efficiently enhance the delivery of B3+ into HeLa cells assisted by an electrostatic interaction-driven anion-exchange process. Fluorescence imaging reveals that B3+ can initially accumulate in the mitochondria after entering HeLa cells, followed by inducing mitochondrial dysfunction and then migrating into the nucleus. Strong B3+ fluorescence translocating from mitochondria to nucleus can be monitored in real-time, allowing for self-reporting of mitochondrial dysfunction in HeLa cells. Moreover, we demonstrate that B3NPs exert significantly higher cytotoxicity against seven different tumor cells (e.g., U87MG, HeLa, MDA-MB-468, MDA-MB-435, MDA-MB-231, MCF-7, and HCT116 cells) compared to human normal tissue cells (e.g., HUVEC, HEK293). This work highlights the utility of the self-assembly approach to improve the cytotoxicity and selectivity of mitochondria-targeting agents against tumor cells.


Asunto(s)
Antineoplásicos/farmacología , Berberina/farmacología , Materiales Biocompatibles/farmacología , Mitocondrias/efectos de los fármacos , Antineoplásicos/química , Berberina/análogos & derivados , Berberina/química , Materiales Biocompatibles/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ensayo de Materiales , Mitocondrias/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Estructura Molecular , Tamaño de la Partícula
19.
Research (Wash D C) ; 2020: 4087069, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33029587

RESUMEN

Noninvasive in vivo imaging of hepatic glutathione (GSH) levels is essential to early diagnosis and prognosis of acute hepatitis. Although GSH-responsive fluorescence imaging probes have been reported for evaluation of hepatitis conditions, the low penetration depth of light in liver tissue has impeded reliable GSH visualization in the human liver. We present a liver-targeted and GSH-responsive trimodal probe (GdNPs-Gal) for rapid evaluation of lipopolysaccharide- (LPS-) induced acute liver inflammation via noninvasive, real-time in vivo imaging of hepatic GSH depletion. GdNPs-Gal are formed by molecular coassembly of a GSH-responsive Gd(III)-based MRI probe (1-Gd) and a liver-targeted probe (1-Gal) at a mole ratio of 5/1 (1-Gd/1-Gal), which shows high r 1 relaxivity with low fluorescence and fluorine magnetic resonance spectroscopic (19F-MRS) signals. Upon interaction with GSH, 1-Gd and 1-Gal are cleaved and GdNPs-Gal rapidly disassemble into small molecules 2-Gd, 2-Gal, and 3, producing a substantial decline in r 1 relaxivity with compensatory enhancements in fluorescence and 19F-MRS. By combining in vivo magnetic resonance imaging (1H-MRI) with ex vivo fluorescence imaging and 19F-MRS analysis, GdNPs-Gal efficiently detect hepatic GSH using three independent modalities. We noninvasively visualized LPS-induced liver inflammation and longitudinally monitored its remediation in mice after treatment with an anti-inflammatory drug, dexamethasone (DEX). Findings highlight the potential of GdNPs-Gal for in vivo imaging of liver inflammation by integrating molecular coassembly with GSH-driven disassembly, which can be applied to other responsive molecular probes for improved in vivo imaging.

20.
ACS Nano ; 13(2): 2544-2557, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30730695

RESUMEN

Combination therapy, especially photodynamic/photothermal therapy (PDT/PTT), has shown promising applications in cancer therapy. However, sequential irradiation by two different laser sources and even the utilization of single high-power laser to induce either combined PDT/PTT or individual PTT will be subjected to prolonged treatment time, complicated treatment process, and potential skin burns. Thus, low power single laser activatable combined PDT/PTT is still a formidable challenge. Herein, we propose an effective strategy to achieve synergistic cancer phototherapy under low power single laser irradiation for short duration. By taking advantage of dual plasmonic PTT nanoagents (AuNRs/MoS2), a significant increase in temperature up to 60 °C with an overall photothermal conversion efficiency (PCE) of 68.8% was achieved within 5 min under very low power (0.2 W/cm2) NIR laser irradiation. The enhanced PCE and PTT performance is attributed to the synergistic plasmonic PTT effect (PPTT) of dual plasmonic nanoagents, promoting simultaneous release (85%) of electrostatically bonded indocyanine green (ICG) to induce PDT effects, offering simultaneous PDT/synergistic PPTT. Both in vitro and in vivo investigations reveal complete cell/tumor eradication, implying that simultaneous PDT/synergistic PPTT effects induced by AuNRs/MoS2-ICG are much superior over individual PDT or synergistic PPTT. Notably, synergistic PPTT induced by dual plasmonic nanoagents also demonstrates higher in vivo antitumor efficacy than either individual PDT or PTT agents. Taken together, under single laser activation with low power density, the proposed strategy of simultaneous PDT/synergistic PPTT effectively reduces the treatment time, achieves high therapeutic index, and offers safe treatment option, which may serve as a platform to develop safer and clinically translatable approaches for accelerating cancer therapeutics.


Asunto(s)
Rayos Láser , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Fototerapia , Proliferación Celular/efectos de los fármacos , Disulfuros/química , Disulfuros/farmacología , Relación Dosis-Respuesta a Droga , Oro/química , Oro/farmacología , Células HeLa , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Verde de Indocianina/química , Verde de Indocianina/farmacología , Molibdeno/química , Molibdeno/farmacología , Imagen Óptica , Fármacos Fotosensibilizantes/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA