Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomed Opt Express ; 15(2): 924-937, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38404313

RESUMO

In vivo near infrared (NIR) fluorescence imaging and laser speckle contrast imaging (LSCI) are emerging optical bioimaging modalities, which can provide information on blood vessels morphology, volume and the blood flow velocity. Optical tissue clearing (OTC) technique addresses a light scattering problem in optical bioimaging, which is imperative for the transcranial brain imaging. Herein, we report an approach combining NIR fluorescence and LSC microscopy imaging with OTC. A liposomal nanoformulation comprising NIR fluorescent dye ICG and photosensitizer BPD was synthesized and injected intravenously into mouse with OTC treated skull. Transcranial excitation of BPD in nanoliposomes resulted in the localized, irradiation dose dependent photodynamic damage of the brain blood vessels, which was manifested both in NIR fluorescence and LSC transcranial imaging, revealing changes in the vessels morphology, volume and the blood flow rate. The developed approach allows for bimodal imaging guided, localized vascular PDT of cancer and other diseases.

2.
J Photochem Photobiol B ; 250: 112816, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38029664

RESUMO

Although photobiomodulation (PBM) and gamma visual stimulatqion (GVS) have been overwhelmingly explored in the recent time as a possible light stimulation (LS) means of Alzheimer's disease (AD) therapy, their effects have not been assessed at once. In our research, the AD mouse model was stimulated using light with various parameters [continuous wave (PBM) or 40 Hz pulsed visible LED (GVS) or 40 Hz pulsed 808 nm LED (PBM and GVS treatment)]]. The brain slices collected from the LS treated AD model mice were evaluated using (i) fluorescence microscopy to image thioflavine-S labeled amy-loid-ß (Aß) plaques (the main hallmark of AD), or (ii) two-photon excited fluorescence (TPEF) imaging of unlabeled Aß plaques, showing that the amount of Aß plaques was reduced after LS treatment. The imaging results correlated well with the results of Morris water maze (MWM) test, which demonstrated that the spatial learning and memory abilities of LS treated mice were noticeably higher than those of untreated mice. The LS effect was also assessed by in vivo nonlinear optical imaging, revealing that the cerebral amyloid angiopathy decreased spe-cifically as a result of 40 Hz pulsed 808 nm irradiation, on the contrary, the angiopathy reversed after visible 40 Hz pulsed light treatment. The obtained results provide useful reference for further optimization of the LS (PBM or GVS) parameters to achieve efficient phototherapy of AD.


Assuntos
Doença de Alzheimer , Terapia com Luz de Baixa Intensidade , Camundongos , Animais , Estimulação Luminosa , Terapia com Luz de Baixa Intensidade/métodos , Encéfalo/metabolismo , Placa Amiloide , Peptídeos beta-Amiloides , Modelos Animais de Doenças , Camundongos Transgênicos
3.
Cytometry A ; 105(4): 252-265, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38038631

RESUMO

Mesenchymal stem cells (MSCs) being injected into the body can stimulate or decelerate carcinogenesis. Here, the direction of influence of human placenta-derived MSCs (P-MSCs) on the Lewis lung carcinoma (LLC) tumor development and metastatic potential is investigated in C57BL/6 mice depending on the injection method. After intramuscular co-inoculation of LLC and P-MSCs (LLC + P-MSCs), the growth of primary tumor and angiogenesis are slowed down compared to the control LLC on the 15th day. This is explained by the fact of a decrease in the secretion of proangiogenic factors during in vitro co-cultivation of an equal amount of LLC and P-MSCs. When P-MSCs are intravenously (i.v.) injected in the mice with developing LLC (LLC + P-MSCs(i.v.)), the tumor growth and angiogenesis are stimulated on the 15th day. A highly activated secretion of proangiogenic factors by P-MSCs in a similar in vitro model can explain this. In both the models compared to the control on the 23rd day, there is no significant difference in the tumor growth, while angiogenesis remains correspondingly decelerated or stimulated. However, in both the models, the total volume and number of lung metastases constantly increase compared to the control: it is mainly due to small-size metastases for LLC + P-MSCs(i.v.) and larger ones for LLC + P-MSCs. The increase in the rate of LLC cell dissemination after the injection of P-MSCs is explained by the disordered polyploidy and chromosomal instability, leading to an increase in migration and invasion of cancer cells. After LLC + P-MSCs co-inoculation, the tumor cell karyotype has the most complex and heterogeneous chromosomal structure. These findings indicate a bidirectional effect of P-MSCs on the growth of LLC in the early periods after injection, depending on the injection method, and, correspondingly, the number of contacting cells. However, regardless of the injection method, P-MSCs are shown to increase LLC aggressiveness related to cancer-associated angiogenesis and metastasis activation in the long term.


Assuntos
Carcinoma Pulmonar de Lewis , Neoplasias Pulmonares , Células-Tronco Mesenquimais , Humanos , Camundongos , Animais , Carcinoma Pulmonar de Lewis/patologia , Camundongos Endogâmicos C57BL , Neoplasias Pulmonares/patologia
4.
Anal Chim Acta ; 1254: 341086, 2023 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-37005018

RESUMO

Plasmon-enhanced luminescence (PEL) is a unique photophysical phenomenon in which the interaction between luminescent moieties and metal nanostructures results in a marked luminescence enhancement. PEL offers several advantages and has been extensively used to design robust biosensing platforms for luminescence-based detection and diagnostics applications, as well as for the development of many efficient bioimaging platforms, enabling high-contrast non-invasive real-time optical imaging of biological tissues, cells, and organelles with high spatial and temporal resolution. This review summarizes recent progress in the development of various PEL-based biosensors and bioimaging platforms for diverse biological and biomedical applications. Specifically, we comprehensively assessed rationally designed PEL-based biosensors that can efficiently detect biomarkers (proteins and nucleic acids) in point-of-care tests, highlighting significant improvements in the sensing performance upon the integration of PEL. In addition to discussing the merits and demerits of recently developed PEL-based biosensors on substrates or in solutions, we include a brief discussion on integrating PEL-based biosensing platforms into microfluidic devices as a promising multi-responsive detection method. The review also presents comprehensive details about the recent advances in the development of various PEL-based multi-functional (passive targeting, active targeting, and stimuli-responsive) bioimaging probes, highlighting the scope of future improvements in devising robust PEL-based nanosystems to achieve more effective diagnostic and therapeutic insights by enabling imaging-guided therapy.


Assuntos
Técnicas Biossensoriais , Nanoestruturas , Luminescência , Nanoestruturas/química , Técnicas Biossensoriais/métodos , Imagem Óptica
5.
J Biophotonics ; 16(7): e202300018, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37021842

RESUMO

Label-free hyperspectral imaging (HSI) of lipids was demonstrated in the near-infrared (NIR) and shortwave infrared (SWIR) regions (950-1800 nm) using porcine tissue. HSI was performed in the transmission light-pass configuration, using a NIR-SWIR camera coupled with a liquid crystal tunable filter. The transmittance spectra of the regions of interest (ROIs), which correspond to the lipid and muscle areas in the specimen, were utilized for the spectrum unmixing. The transmittance spectra in ROIs were compared with those recorded by a spectrophotometer using samples of adipose and muscle. The lipid optical absorption bands at 1210 and 1730 nm were first used for the unmixing and mapping. Then, we performed the continuous multiband unmixing over the entire available spectral range, thereby, considering a combination of characteristic absorption bands of lipids, proteins, and water. The enhanced protocol demonstrates the ability to visualize small adipose inclusions of 1-10 µm size.


Assuntos
Imageamento Hiperespectral , Água , Animais , Suínos , Projetos Piloto , Músculos/diagnóstico por imagem , Obesidade , Lipídeos
6.
Small ; 19(8): e2205165, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36508710

RESUMO

The conventional approach in cancer nanomedicine involves advanced drug nanocarriers delivering preloaded therapeutics to targeted tumor sites to maximize drug efficiency. However, both cancer drugs and nanocarriers inevitably produce side effects and systemic toxicity. Herein, hemoglobin nanocrystals (HbC) as drug-free theranostic nanoformulations with the tumor microenvironment (TME) activated diagnostic and therapeutic abilities towards colon tumors are introduced. HbC can release Fe2+ oxidized to Fe3+ in the Fenton reaction with tumor endogenous H2 O2 , concurrently with the generation of cytotoxic hydroxyl radicals (•OH) that allow for chemodynamic therapy (CDT). Furthermore, in situ-produced Fe3+ reacts with colon tumor-abundant H2 S, resulting in the production of Fe1- x S, which provides magnetic resonance imaging (MRI) contrast and allows for NIR light-inducible photothermal therapy (PTT). In vitro and in vivo studies revealed that HbC produced CDT towards 4T1 tumors, and MRI-guided, synergistically enhanced combination of CDT and PTT against H2 S abundant colon tumors (CT26), with negligible toxicity towards normal tissues, enlightening HbC as highly efficient and biocompatible TME activated theranostic nanoplatform specific against colon cancer without any traditional drugs and drug carriers.


Assuntos
Antineoplásicos , Neoplasias do Colo , Nanopartículas , Neoplasias , Humanos , Linhagem Celular Tumoral , Medicina de Precisão , Nanopartículas/química , Antineoplásicos/farmacologia , Neoplasias/terapia , Neoplasias do Colo/tratamento farmacológico , Microambiente Tumoral , Nanomedicina Teranóstica , Peróxido de Hidrogênio/farmacologia
7.
Nanoscale ; 14(39): 14770-14778, 2022 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-36178268

RESUMO

Lanthanide-doped fluoride nanocrystals (NCs) are known to exhibit unique optical properties, such as upconversion and downconversion luminescence (UCL and DCL), which can be employed for various applications. In this work, we demonstrate that by doping praseodymium(III) and ytterbium(III) ions (Pr3+ and Yb3+) into a nanosized fluoride matrix (i.e. NaYF4 and LiYF4), it is possible to combine their UCL and DCL properties that can be concurrently used for biomedical applications. In particular, the emissive modes combined in a single nanoparticle co-doped with Pr3+ and Yb3+ include DCL emission (excited at 980 nm and peaked at 1320 nm), which can be used for near infrared (NIR) DCL bioimaging in the NIR-II window of biological tissue transparency (∼1000-1350 nm) and UCL emission (excited at 447 nm and peaked at 275 nm) that can be employed for germicide action (via irradiation by light in the UVC range). A possibility of the latter was demonstrated by the denaturation of double-stranded DNA (dsDNA) into single-stranded ones that was caused by the UVC UCL emission from the NCs under 447 nm irradiation; it was evidenced by the hyperchromicity observed in the irradiated dsDNA solution and also by a fluorometric analysis of DNA unwinding (FADU) assay. Concurrently, the possibility of NIR-II luminescence bioimaging through biological tissues (bovine tooth and chicken flesh) was demonstrated. The proposed concept paves a way for NIR-II imaging guided antimicrobial phototherapy using lanthanide-doped fluoride nanocrystals.


Assuntos
Elementos da Série dos Lantanídeos , Nanopartículas , Animais , Bovinos , DNA , Fluoretos/química , Elementos da Série dos Lantanídeos/química , Luminescência , Nanopartículas/química , Praseodímio , Itérbio/química
8.
Alzheimers Res Ther ; 14(1): 84, 2022 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-35717405

RESUMO

BACKGROUND: Low-intensity light can decelerate neurodegenerative disease progression and reduce amyloid ß (Aß) levels in the cortex, though the cellular and molecular mechanisms by which photobiomodulation (PBM) protects against neurodegeneration are still in the early stages. Microglia cells play a key role in the pathology of Alzheimer's disease by causing chronic inflammation. We present new results concerning the PBM of both oxidative stress and microglia metabolism associated with the activation of metabolic processes by 808 nm near-infrared light. METHODS: The studies were carried out using healthy male mice to obtain the microglial cell suspension from the hippocampus. Oligomeric ß-amyloid (1-42) was prepared and used to treat microglia cells. Light irradiation of cells was performed using diode lasers emitting at 808 nm (30 mW/cm2 for 5 min, resulting in a dose of 10 J/cm2). Mitochondrial membrane potential, ROS level studies, cell viability, apoptosis, and necrosis assays were performed using epifluorescence microscopy. Phagocytosis, nitric oxide and H2O2 production, arginase, and glucose 6-phosphate dehydrogenase activities were measured using standard assays. Cytokines, glucose, lactate, and ATP were measurements with ELISA. As our data were normally distributed, two-way ANOVA test was used. RESULTS: The light induces a metabolic shift from glycolysis to mitochondrial activity in pro-inflammatory microglia affected by oligomeric Aß. Thereby, the level of anti-inflammatory microglia increases. This process is accompanied by a decrease in pro-inflammatory cytokines and an activation of phagocytosis. Light exposure decreases the Aß-induced activity of glucose-6-phosphate dehydrogenase, an enzyme that regulates the rate of the pentose phosphate pathway, which activates nicotinamide adenine dinucleotide phosphate oxidases to further produce ROS. During co-cultivation of neurons with microglia, light prevents the death of neurons, which is caused by ROS produced by Aß-altered microglia. CONCLUSIONS: These original data clarify reasons for how PBM protects against neurodegeneration and support the use of light for therapeutic research in the treatment of Alzheimer's disease.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Citocinas/metabolismo , Glucose/metabolismo , Humanos , Peróxido de Hidrogênio , Masculino , Camundongos , Microglia/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Fototerapia , Espécies Reativas de Oxigênio/metabolismo
9.
Artigo em Inglês | MEDLINE | ID: mdl-35549055

RESUMO

Rational design of tumor-microenvironment (TME)-activated nanoformulation for precisely targeted cancer treatment has recently attracted an enormous attention. However, the all-in-one TME-activated theranostic nanosystems with a simple preparation and high biocompatibility are still rarely reported. Herein, catalase nanocrystals (CatCry) are first introduced as a tumor microenvironment activatable nanoplatform for selective theranostics of colon cancer. They are engaged as (i) a "nanoreactor" for silver nanoparticles (AgNP) synthesis, (ii) a nanovehicle for tumor delivery of anticancer drug doxorubicin (DOX), and (iii) an in situ O2 generator to relief tumor hypoxia. When CatCry-AgNP-DOX nanoformulation is within a tumor, the intratumoral H2S turns AgNP into Ag2S nanoparticles, inducing a photothermal effect and NIR-II emission under 808 nm laser irradiation and also triggering DOX release. Simultaneously, CatCry catalyzes intratumoral H2O2 into O2, relieving hypoxia and enhancing chemotherapy. In contrast, when delivered to healthy tissue without increased concentration of H2S, the developed nanoformulation remains in the "off" state and no theranostic action takes place. Studies with colon cancer cells in vitro and a murine colon cancer model in vivo demonstrated that CatCry-AgNP-DOX delivered a synergistic combination of PTT and enhanced chemotherapy, enabling complete eradication of tumor with minimal side effects. This work not only introduces nanoplatform for theranostics of H2S-rich tumors but also suggests a general strategy for protein-crystal-based nanomedicine.

10.
J Photochem Photobiol B ; 227: 112388, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35074677

RESUMO

Irradiation with red or near-infrared (NIR) light in low level light therapy (LLLT) is found to stimulate cellular processes and bioenergetics, resulting in enhanced wound healing, pain control, neurodegenerative diseases treatment, etc. During light irradiation of tissues and organs, different cells are affected, though the connection between photostimulation of cells and their environmental conditions remains poorly understood. In this report, red/NIR light-stimulated angiogenesis is investigated using endothelial cells in vitro, with a focus on the capillary-like structure (CLS) formation and the respective biochemical processes in cells under conditions proximate to a healthy or malignant environment, which strongly defines angiogenesis. To model environmental conditions for endotheliocytes in vitro, the cell culture environment was supplemented by an augmented conditioned medium from macrophages or cancer cells. The biochemical processes in endothelial cell cultures were investigated with and without irradiation by red (650 nm) and near-infrared (808 nm) laser diodes and under normoxia or hypoxia conditions. A light-stimulated angiogenesis has been found, with a more efficient stimulation by 650 nm light compared to 808 nm light. It was shown that the irradiation with light promoted extracellular Ca2+ influx, fostered cell cycle progression, proliferation and NO generation in endothelial cells, and caused an increase in vascular endothelial growth factor (VEGF) production by endothelial cells and M2 macrophages under hypoxia conditions. The activation of VEGF production by macrophages was found to be associated with an increase in the number of M2 macrophages after light irradiation under hypoxia conditions. Thus, a new pathway of an activation of the endothelial cell metabolism, which is related with the extracellular Ca2+ influx after light irradiation, has been revealed. STATEMENT OF SIGNIFICANCE: Red/NIR light-stimulated angiogenesis has been studied using endothelial cells in vitro, with focus on CLS formation and the respective biochemical processes in cell models proximate to a healthy or malignant environment. A light-stimulated angiogenesis has been found, stimulated via extracellular Ca2+ influx, cell cycle progression, proliferation and NO generation, VEGF production increase by endothelial cells under hypoxia conditions.


Assuntos
Células Endoteliais , Fator A de Crescimento do Endotélio Vascular , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Raios Infravermelhos , Macrófagos/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G142-G153, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34851733

RESUMO

Hepatocellular carcinoma (HCC) is the most common primary liver cancer, which is not sensitive to radiotherapy and chemotherapy and very often experiences postoperative relapse. In this regard, effective screening of liver cancer is considered as the most important and urgent task. The aim of our study was to determine whether N-methyl-D-aspartate receptor (NMDAR) and, in particular, its subunits, can serve as biomarkers to distinguish the precancerous liver at early stages of liver fibrosis. We assessed the development of HCC after 10, 15, and 22 wk using a HCC rat model. The expression of NMDAR subunits was monitored at different stages of HCC by means of immunohistochemistry combined with epifluorescence microscopy imaging, Western blotting, and direct bisulfite sequencing. NMDAR subunits were not found in healthy liver tissues. In contrast, NMDAR subunits, in particular NR1 and NR2B, appeared at the stage of severe liver fibrosis (precancerous liver disease) in rats and were expressed during the development of HCC in rats and mice. Using the direct bisulfite sequencing, we detected that increased expression of NMDAR directly correlated with the demethylation of CpG islands in the promoter region of genes encoding receptor subunits. The obtained results confirmed that NMDAR subunits can serve as new biomarkers of precancerous liver disease, severe fibrosis, and its progression towards HCC.NEW & NOTEWORTHY We have shown NMDAR expression in cell transformation process at early stages of cancer, specifically HCC. The aim of our study was to define the disease stages from precancerous liver disease towards liver cancer progression when NMDAR subunits were expressed/detected. A fibrosis/HCC rat model, immunohistochemistry combined with epifluorescence microscopy imaging, Western blotting was used. The dynamics of appearance of NMDAR subunits, their expression and methylation status during the development of HCC were shown and discussed.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/fisiologia , Animais , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , RNA Mensageiro/metabolismo , Ratos , Roedores/genética , Roedores/metabolismo
12.
Photochem Photobiol ; 98(2): 484-497, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34569637

RESUMO

Low-level light therapy (LLLT) is emerging as a promising therapeutic approach to modulate the biochemical and molecular processes within living cells. LLLT is known to produce local and systemic effects; therefore, immune cells in local tissues or in the circulation are affected by light. However, this specific effect remains weakly explored. In this study, the effect of red (650 nm) and NIR (808 nm) light on phagocytosis (respiratory burst), cytokine expression, mitochondrial activity, ROS generation, Ca2+ influx and membrane depolarization in macrophages in vitro is investigated. Both the phagocytic capacity and adhesion of macrophages strongly (~2.5 times) increased in the first hours after exposure to light in a dose-dependent manner. The light-evoked upregulation of phagocytosis is found to be less efficient than the maximal pharmacologically induced enhancement of ~3.2 times. Also, red/NIR light reduces the production of pro-inflammatory cytokines and activates the secretion of anti-inflammatory cytokines by several times in activated macrophages. At the same time, the viability shows a biphasic dose response: it increases after irradiation with lower doses (0.3-1 J cm-2 ) and decreases after treatment with higher doses (18-30 J cm-2 ), which is apparently associated with the upregulation of ROS generation, followed by an increase in the mitochondrial activity.


Assuntos
Cálcio/metabolismo , Citocinas , Terapia com Luz de Baixa Intensidade , Citocinas/metabolismo , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Fagocitose , Espécies Reativas de Oxigênio/metabolismo
13.
Small ; 17(41): e2103569, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34532978

RESUMO

Photodynamic therapy (PDT) is a well-known method for cancer therapy in the clinic. However, the inherent hypoxia microenvironment of solid tumors enormously restricts the PDT efficiency. Herein, catalase nanocrystals (CatCry) are introduced as in situ oxygen (O2 )-generating system to relieve tumor hypoxia and enhance PDT efficiency for solid tumors. After loading with photosensitizer methylene blue (MB), a PDT drug platform (CatCry-MB) emerges, allowing for significant increasing PDT efficiency instigated by three factors. First, the high stability and recyclable catalytic activity of CatCry enable a long-term endogenous H2 O2 decomposition for continuous O2 supply for sustained relief of tumor hypoxia. Second, both the produced O2 and loaded MB are confined within CatCry nanoporous structure, shortening the diffusion distance between O2 and MB to maximize the production of singlet oxygen (1 O2 ). Third, the MB molecules are uniformly dispersed within CatCry lattice, avoiding MB aggregation and causing more MB molecules be activated to produce more 1 O2 . With the three complementary mechanisms, tumor hypoxia is eradicated and the resulted enhancement in PDT efficiency is demonstrated in vitro and in vivo. The proposed approach opens up a new venue for the development of other O2 -dependent tumor treatments, such as chemotherapy, radiotherapy, and immunotherapy.


Assuntos
Nanopartículas , Fotoquimioterapia , Catalase , Linhagem Celular Tumoral , Humanos , Hipóxia/tratamento farmacológico , Azul de Metileno , Oxigênio , Fármacos Fotossensibilizantes/uso terapêutico
14.
Biosensors (Basel) ; 11(8)2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34436057

RESUMO

Alzheimer's disease (AD) is a multifactorial, irreversible, and incurable neurodegenerative disease. The main pathological feature of AD is the deposition of misfolded ß-amyloid protein (Aß) plaques in the brain. The abnormal accumulation of Aß plaques leads to the loss of some neuron functions, further causing the neuron entanglement and the corresponding functional damage, which has a great impact on memory and cognitive functions. Hence, studying the accumulation mechanism of Aß in the brain and its effect on other tissues is of great significance for the early diagnosis of AD. The current clinical studies of Aß accumulation mainly rely on medical imaging techniques, which have some deficiencies in sensitivity and specificity. Optical imaging has recently become a research hotspot in the medical field and clinical applications, manifesting noninvasiveness, high sensitivity, absence of ionizing radiation, high contrast, and spatial resolution. Moreover, it is now emerging as a promising tool for the diagnosis and study of Aß buildup. This review focuses on the application of the optical imaging technique for the determination of Aß plaques in AD research. In addition, recent advances and key operational applications are discussed.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doenças Neurodegenerativas , Doença de Alzheimer/diagnóstico por imagem , Humanos , Imagem Óptica , Placa Amiloide/diagnóstico por imagem
15.
Front Chem ; 9: 659553, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33981673

RESUMO

Rare-earth doped nanoparticles (RENPs) have been widely used for anti-counterfeiting and security applications due to their light frequency conversion features: they are excited at one wavelength, and they display spectrally narrow and distinguished luminescence peaks either at shorter wavelengths (i.e., frequency/energy upconversion) or at longer wavelengths (frequency/energy downconversion). RENPs with a downconversion (DC) photoluminescence (PL) in short-wave infrared (SWIR) spectral range (~1,000-1,700 nm) have recently been introduced to anti-counterfeiting applications, allowing for multilevel protection based on PL imaging through opaque layers, due to a lesser scattering of SWIR PL emission. However, as the number and spectral positions of the discrete PL bands exhibited by rare-earth ions are well-known, it is feasible to replicate luminescence spectra from RENPs, which results in a limited anti-counterfeiting security. Alternatively, lifetime of PL from RENPs can be used for encoding, as it can be finely tuned in broad temporal range (i.e., from microseconds to milliseconds) by varying type of dopants and their content in RENPs, along with the nanoparticle morphology and size. Nevertheless, the current approach to decoding and imaging the RENP luminescence lifetimes requires multiple steps and is highly time-consuming, precluding practical applications of PL lifetime encoding for anti-counterfeiting. Herein, we report the use of a rapid lifetime determination (RLD) technique to overcome this issue and introduce real-time imaging of SWIR PL lifetime for anti-counterfeiting applications. NaYF4:20% Yb, x% Er (x = 0, 2, 20, 80)@NaYF4 core@shell RENPs were synthesized and characterized, revealing DC PL in SWIR region, with maximum at ~1,530 nm and PL lifetimes ranging from 3.2 to 6 ms. Imaging of the nanoparticles with different lifetimes was performed by the developed time-gated imaging system engaging RLD method and the precise manipulation of the delay between the excitation pulses and camera gating windows. Moreover, it is shown that imaging and decrypting can be performed at a high rate (3-4 fps) in a cyclic manner, thus allowing for real-time temporal decoding. We believe that the demonstrated RLD-based fast PL lifetime imaging approach can be employed in other applications of photoluminescent RENPs.

16.
Nanomaterials (Basel) ; 11(5)2021 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-33923050

RESUMO

Magneto-plasmonic nanocomposites can possess properties inherent to both individual components (iron oxide and gold nanoparticles) and are reported to demonstrate high potential in targeted drug delivery and therapy. Herein, we report on Fe3O4/Au magneto-plasmonic nanocomposites (MPNC) synthesized with the use of amino acid tryptophan via chemical and photochemical reduction of Au ions in the presence of nanosized magnetite. The magnetic field (MF) induced aggregation was accompanied by an increase in the absorption in the near-infrared (NIR) spectral region, which was demonstrated to provide an enhanced photothermal (PT) effect under NIR laser irradiation (at 808 nm). A possibility for therapeutic application of the MPNC was illustrated using cancer cells in vitro. Cultured HeLa cells were treated by MPNC in the presence of MF and without it, following laser irradiation and imaging using confocal laser scanning microscopy. After scanning laser irradiation of the MPNC/MF treated cells, a formation and rise of photothermally-induced microbubbles on the cell surfaces was observed, leading to a damage of the cell membrane and cell destruction. We conclude that the synthesized magneto-plasmonic Fe3O4/Au nanosystems exhibit magnetic field-induced reversible aggregation accompanied by an increase in NIR absorption, allowing for an opportunity to magnetophoretically control and locally enhance a NIR light-induced thermal effect, which holds high promise for the application in photothermal therapy.

17.
Cancer Nanotechnol ; 12(1): 4, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33603920

RESUMO

BACKGROUND: In this study, we report on the synthesis, imaging, and radiosensitizing properties of ultrasmall ß-NaGdF4:Yb50% nanoparticles as a multifunctional theranostic platform. The synthesized nanoparticles act as potent bimodal contrast agents with superior imaging properties compared to existing agents used for magnetic resonance imaging (MRI) and computed tomography (CT). Clonogenic assays demonstrated that these nanoparticles can act as effective radiosensitizers, provided that the nanoparticles are taken up intracellularly. RESULTS: Our ultrasmall ß-NaGdF4:Yb50% nanoparticles demonstrate improvement in T1-weighted contrast over the standard clinical MR imaging agent Gd-DTPA and similar CT signal enhancement capabilities as commercial agent iohexol. A 2 Gy dose of X-ray induced ~ 20% decrease in colony survival when C6 rat glial cells were incubated with non-targeted nanoparticles (NaGdF4:Yb50%), whereas the same X-ray dose resulted in a ~ 60% decrease in colony survival with targeted nanoparticles conjugated to folic acid (NaGdF4:Yb50%-FA). Intravenous administration of nanoparticles resulted in clearance through urine and feces within a short duration, based on the ex vivo analysis of Gd3+ ions via ICP-MS. CONCLUSION: These biocompatible and in vivo clearable ultrasmall NaGdF4:Yb50% are promising candidates for further evaluation in image-guided radiotherapy applications.

18.
Biomed Opt Express ; 12(12): 7544-7555, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-35003851

RESUMO

The blood-brain barrier (BBB) is a dynamic barrier between the blood microcirculation system and the brain parenchyma, which plays an important role in the pathogenesis of a variety of neurological diseases. Meanwhile, a non-invasive therapeutic approach of photobiomodulation (PBM) has emerged as a promising treatment for neurological disorders through irradiation with near infrared (NIR) light. However, despite multiple encouraging results reported for PBM in vitro and in vivo, the mechanisms of its therapeutic effect on brain, especially on the BBB, remain barely known. Herein, the effect of NIR light irradiation on the in vitro BBB models was studied. 808 nm laser irradiation at the doses of 10 and 30 J/cm2 was found to significantly increase the permeability of this BBB model. The results showed that NIR light affected mitochondria of cells in the in vitro BBB models, leading to an increase in the mitochondrial activity, reactive oxygen species (ROS) level and Ca2+ influx. The activity of matrix metalloproteinases and the expression of the tight junction proteins in the endothelial cells were found to be inhibited by the NIR light, resulting in an increase in the BBB permeability. This study suggested a new strategy for drug transport across the BBB in development of treatments for brain disorders.

19.
J Photochem Photobiol B ; 214: 112088, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33278762

RESUMO

Low level light therapy uses light of specific wavelengths in red and near-infrared spectral range to treat various pathological conditions. This light is able to modulate biochemical cascade reactions in cells that can have important health implications. In this study, the effect of low intensity light at 650, 808 and 1064 nm on neurons and two types of cancer cells (neuroblastoma and HeLa) is reported, with focus on the photoinduced change of intracellular level of Ca2+ ions and corresponding signaling pathways. The obtained results show that 650 and 808 nm light promotes intracellular Ca2+ elevation regardless of cell type, but with different dynamics due to the specificities of Ca2+ regulation in neurons and cancer cells. Two origins responsible for Ca2+ elevation are determined to be: influx of exogenous Ca2+ ions into cells and Ca2+ release from endoplasmic reticulum. Our investigation of the related cellular processes shows that light-induced membrane depolarization is distinctly involved in the mechanism of Ca2+ influx. Ca2+ release from endoplasmic reticulum activated by reactive oxygen species generation is considered as a possible light-dependent signaling pathway. In contrast to the irradiation with 650 and 808 nm light, no effects are observed under 1064 nm irradiation. We believe that the obtained insights are of high significance and can be useful for the development of drug-free phototherapy.


Assuntos
Sinalização do Cálcio/efeitos da radiação , Cálcio/efeitos da radiação , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/efeitos da radiação , Cálcio/fisiologia , Membrana Celular/metabolismo , Eletrofisiologia , Corantes Fluorescentes/química , Células HeLa , Humanos , Raios Infravermelhos , Terapia com Luz de Baixa Intensidade , Neurônios/efeitos da radiação , Imagem Óptica , Espécies Reativas de Oxigênio/efeitos da radiação
20.
Colloids Surf B Biointerfaces ; 198: 111458, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33246782

RESUMO

Ovarian cancer (OVC), the most lethal form of all gynecological cancers, is a big threat to women's health. Late diagnosis at the advanced stages is one of the major reasons for the ovarian cancer-related deaths. Conventionally, the up-regulated proteins CA125 (cancer antigen 125) and HE4 (human epididymis protein 4) are used as biomarkers to diagnose the OVC malignancies. The lack of sensitivity/specificity and the false-positive results create complexity in the diagnostic process. With specificity over 90 %, HE4 is suitable for diagnosing ovarian cancer. Herein, we have developed an ultrasensitive all-graphene quantum dot (GQD) Förster resonance energy transfer (FRET) probe for the ratiometric detection of HE4 biomarker. A set of two GQD samples were solvothermally prepared and then analyzed by the morphological, structural, and photophysical characterization. One GQD sample exhibited a strong green emission, peaked at around 515 nm, while the other GQD sample displayed a strong red emission with maximum at around 615 nm. The good spectral overlap between the emission and excitation spectra of the green and red GQDs, respectively, all allowed us to consider them for the design of FRET-based probe. The green and red-emitting GQDs were conjugated with HE4 antibody and used as donor and acceptor, respectively for the ratiometric sensing of HE4 ovarian cancer biomarker. The all GQD FRET probe was able to detect as low as 4.8 pM, along with a large dynamic detection range up to 300 nM. The selectivity and interference effect of the developed FRET probe was also investigated against different protein combinations.


Assuntos
Grafite , Neoplasias Ovarianas , Pontos Quânticos , Biomarcadores Tumorais , Feminino , Transferência Ressonante de Energia de Fluorescência , Humanos , Neoplasias Ovarianas/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...