Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 186, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013474

RESUMEN

Organic near-infrared room temperature phosphorescence materials have unparalleled advantages in bioimaging due to their excellent penetrability. However, limited by the energy gap law, the near-infrared phosphorescence materials (>650 nm) are very rare, moreover, the phosphorescence lifetimes of these materials are very short. In this work, we have obtained organic room temperature phosphorescence materials with long wavelengths (600/657-681/732 nm) and long lifetimes (102-324 ms) for the first time through the guest-host doped strategy. The guest molecule has sufficient conjugation to reduce the lowest triplet energy level and the host assists the guest in exciton transfer and inhibits the non-radiative transition of guest excitons. These materials exhibit good tissue penetration in bioimaging. Thanks to the characteristic of long lifetime and long wavelength emissive phosphorescence materials, the tumor imaging in living mice with a signal to background ratio value as high as 43 is successfully realized. This work provides a practical solution for the construction of organic phosphorescence materials with both long wavelengths and long lifetimes.


Asunto(s)
Colorantes Fluorescentes/síntesis química , Sustancias Luminiscentes/síntesis química , Ganglios Linfáticos/diagnóstico por imagen , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Imagen Óptica/métodos , Animales , Benzofenonas/química , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/farmacocinética , Sustancias Luminiscentes/análisis , Sustancias Luminiscentes/farmacocinética , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Pirenos/química , Piridinas/química , Espectroscopía Infrarroja Corta
2.
Theranostics ; 11(15): 7439-7449, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34158859

RESUMEN

X-ray excited persistent luminescence (XEPL) imaging has attracted increasing attention in biomedical imaging due to elimination of autofluorescence, high signal-to-noise ratio and repeatable activation with high penetration. However, optical imaging still suffers from limited for high spatial resolution. Methods: Herein, we report Mn3+-rich manganese oxide (MnOx)-coated chromium-doped zinc gallogermanate (ZGGO) nanoparticles (Mn-ZGGOs). Enhanced XEPL and magnetic resonance (MR) imaging were investigated by the decomposition of MnOx shell in the environment of tumors. We also evaluated the tumor cell-killing mechanism by detection of reactive oxygen (ROS), lipid peroxidation and mitochondrial membrane potential changes in vitro. Furthermore, the in vivo biodistribution, imaging and therapy were studied by U87MG tumor-bearing mice. Results: In the tumor region, the MnOx shell is quickly decomposed to produce Mn3+ and oxygen (O2) to directly generate singlet oxygen (1O2). The resulting Mn2+ transforms endogenous H2O2 into highly toxic hydroxyl radical (·OH) via a Fenton-like reaction. The Mn2+ ions and ZGGOs also exhibit excellent T1-weighted magnetic resonance (MR) imaging and ultrasensitive XEPL imaging in tumors. Conclusion: Both the responsive dual-mode imaging and simultaneous self-supplied O2 for the production of 1O2 and oxygen-independent ·OH in tumors allow for more accurate diagnosis of deep tumors and more efficient inhibition of tumor growth without external activation energy.


Asunto(s)
Radical Hidroxilo/metabolismo , Sustancias Luminiscentes , Compuestos de Manganeso , Nanopartículas , Neoplasias Experimentales , Imagen Óptica , Óxidos , Oxígeno Singlete/metabolismo , Animales , Línea Celular Tumoral , Humanos , Sustancias Luminiscentes/química , Sustancias Luminiscentes/farmacocinética , Sustancias Luminiscentes/farmacología , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacocinética , Compuestos de Manganeso/farmacología , Ratones , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Óxidos/química , Óxidos/farmacocinética , Óxidos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Int J Mol Sci ; 21(16)2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32824188

RESUMEN

Reporter genes are used to visualize intracellular biological phenomena, including viral infection. Here we demonstrate bioluminescent imaging of viral infection using the NanoBiT system in combination with intraperitoneal injection of a furimazine analogue, hydrofurimazine. This recently developed substrate has enhanced aqueous solubility allowing delivery of higher doses for in vivo imaging. The small high-affinity peptide tag (HiBiT), which is only 11 amino-acids in length, was engineered into a clinically used oncolytic adenovirus, and the complementary large protein (LgBiT) was constitutively expressed in tumor cells. Infection of the LgBiT expressing cells with the HiBiT oncolytic virus will reconstitute NanoLuc in the cytosol of the cell, providing strong bioluminescence upon treatment with substrate. This new bioluminescent system served as an early stage quantitative viral transduction reporter in vitro and also in vivo in mice, for longitudinal monitoring of oncolytic viral persistence in infected tumor cells. This platform provides novel opportunities for studying the biology of viruses in animal models.


Asunto(s)
Furanos/farmacocinética , Imidazoles/farmacocinética , Sustancias Luminiscentes/farmacocinética , Proteínas Luminiscentes/genética , Imagen Óptica/métodos , Pirazinas/farmacocinética , Virosis/diagnóstico por imagen , Adenoviridae/genética , Animales , Línea Celular Tumoral , Furanos/administración & dosificación , Células HEK293 , Humanos , Imidazoles/administración & dosificación , Inyecciones Intraperitoneales , Sustancias Luminiscentes/administración & dosificación , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oligopéptidos/genética , Oligopéptidos/metabolismo , Virus Oncolíticos/genética , Pirazinas/administración & dosificación , Proteínas Recombinantes/genética
4.
Inorg Chem ; 59(14): 10285-10303, 2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32633531

RESUMEN

The established platinum-based drugs form covalent DNA adducts to elicit their cytotoxic response. Although they are widely employed, these agents cause toxic side-effects and are susceptible to cancer-resistance mechanisms. To overcome these limitations, alternative metal complexes containing the rhenium(I) tricarbonyl core have been explored as anticancer agents. Based on a previous study ( Chem. Eur. J. 2019, 25, 9206), a series of highly active tricarbonyl rhenium isonitrile polypyridyl (TRIP) complexes of the general formula fac-[Re(CO)3(NN)(ICN)]+, where NN is a chelating diimine and ICN is an isonitrile ligand, that induce endoplasmic reticulum (ER) stress via activation of the unfolded protein response (UPR) pathway are investigated. A total of 11 of these TRIP complexes were synthesized, modifying both the equatorial polypyridyl and axial isonitrile ligands. Complexes with more electron-donating equatorial ligands were found to have greater anticancer activity, whereas the axial ICN ligands had a smaller effect on their overall potency. All 11 TRIP derivatives trigger a similar phenotype that is characterized by their abilities to induce ER stress and activate the UPR. Lastly, we explored the in vivo efficacy of one of the most potent complexes, fac-[Re(CO)3(dmphen)(ptolICN)]+ (TRIP-1a), where dmphen = 2,9-dimethyl-1,10-phenanthroline and ptolICN = para-tolyl isonitrile, in mice. The 99mTc congener of TRIP-1a was synthesized, and its biodistribution in BALB/c mice was investigated in comparison to the parent Re complex. The results illustrate that both complexes have similar biodistribution patterns, suggesting that 99mTc analogues of these TRIP complexes can be used as diagnostic partner agents. The in vivo antitumor activity of TRIP-1a was then investigated in NSG mice bearing A2780 ovarian cancer xenografts. When administered at a dose of 20 mg/kg twice weekly, this complex was able to inhibit tumor growth and prolong mouse survival by 150% compared to the vehicle control cohort.


Asunto(s)
Antineoplásicos/uso terapéutico , Complejos de Coordinación/uso terapéutico , Sustancias Luminiscentes/uso terapéutico , Nitrilos/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Complejos de Coordinación/síntesis química , Complejos de Coordinación/farmacocinética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Células HeLa , Humanos , Ligandos , Sustancias Luminiscentes/síntesis química , Sustancias Luminiscentes/farmacocinética , Ratones Endogámicos BALB C , Nitrilos/síntesis química , Nitrilos/farmacocinética , Renio/química , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/farmacocinética , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Respuesta de Proteína Desplegada/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Nanobiotechnology ; 18(1): 85, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32503549

RESUMEN

BACKGROUND: The unique upconversion properties of rare-earth-doped nanoparticles offers exciting opportunities for biomedical applications, in which near-IR remote activation of biological processes is desired, including in vivo bioimaging, optogenetics, and light-based therapies. Tuning of upconversion in purposely designed core-shell nanoparticles gives access to biological windows in biological tissue. In recent years there have been several reports on NIR-excitable upconverting nanoparticles capable of working in biological mixtures and cellular settings. Unfortunately, most of these nanosystems are based on ytterbium's upconversion at 980 nm, concurrent with water's absorption within the first biological window. Thus, methods to produce robust upconverting nanoplatforms that can be efficiently excited with other than 980 nm NIR sources, such as 808 nm and 1064 nm, are required for biomedical applications. RESULTS: Herein, we report a synthetic method to produce aqueous stable upconverting nanoparticles that can be activated with 808 nm excitation sources, thus avoiding unwanted heating processes due to water absorbance at 980 nm. Importantly, these nanoparticles, once transferred to an aqueous environment using an amphiphilic polymer, remain colloidally stable for long periods of time in relevant biological media, while keeping their photoluminescence properties. The selected polymer was covalently modified by click chemistry with two FDA-approved photosensitizers (Rose Bengal and Chlorin e6), which can be efficiently and simultaneously excited by the light emission of our upconverting nanoparticles. Thus, our polymer-functionalization strategy allows producing an 808 nm-activable photodynamic nanoplatform. These upconverting nanocomposites are preferentially stored in acidic lysosomal compartments, which does not negatively affect their performance as photodynamic agents. Upon 808 nm excitation, the production of reactive oxidative species (ROS) and their effect in mitochondrial integrity were demonstrated. CONCLUSIONS: In summary, we have demonstrated the feasibility of using photosensitizer-polymer-modified upconverting nanoplatforms that can be activated by 808 nm light excitation sources for application in photodynamic therapy. Our nanoplatforms remain photoactive after internalization by living cells, allowing for 808 nm-activated ROS generation. The versatility of our polymer-stabilization strategy promises a straightforward access to other derivatizations (for instance, by integrating other photosensitizers or homing ligands), which could synergistically operate as multifunctional photodynamic platforms nanoreactors for in vivo applications.


Asunto(s)
Nanopartículas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Polímeros , Química Clic , Estabilidad de Medicamentos , Células HeLa , Humanos , Rayos Infrarrojos , Espacio Intracelular/química , Sustancias Luminiscentes/química , Sustancias Luminiscentes/farmacocinética , Sustancias Luminiscentes/efectos de la radiación , Nanopartículas/química , Nanopartículas/metabolismo , Nanopartículas/efectos de la radiación , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/efectos de la radiación , Polímeros/química , Polímeros/farmacocinética , Polímeros/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo
6.
ACS Nano ; 13(11): 12577-12590, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31657911

RESUMEN

In vivo monitoring of cargo protein delivery is critical for understanding the pharmacological efficacies and mechanisms during cancer therapy, but it still remains a formidable challenge because of the difficulty in observing nonfluorescent proteins at high resolution and sensitivity. Here we report an outer-frame-degradable nanovehicle featuring near-infrared (NIR) dual luminescence for real-time tracking of protein delivery in vivo. Upconversion nanoparticles (UCNPs) and fluorophore-doped degradable macroporous silica (DS) with spectral overlap were coupled to form a core-shell nanostructure as a therapeutic protein nanocarrier, which was eventually enveloped with a hyaluronic acid (HA) shell to prevent protein leakage and for recognizing tumor sites. The DS layer served as both a container to accommodate the therapeutic proteins and a filter to attenuate upconversion luminescence (UCL) of the inner UCNPs. After the nanovehicles selectively accumulated at tumor sites and entered cancer cells, intracellular hyaluronidase (HAase) digested the outermost HA protective shell and initiated the outer frame degradation-induced protein release and UCL restoration of UCNPs in the intracellular environment. Significantly, the biodistribution of the nanovehicles can be traced at the 710 nm NIR fluorescence channel of DS, whereas the protein release can be monitored at the 660 nm NIR fluorescence channel of UCNPs. Real-time tracking of protein delivery and release was achieved in vitro and in vivo by NIR fluorescence imaging. Moreover, in vitro and in vivo studies manifest that the protein cytochrome c-loaded nanovehicles exhibited excellent cancer therapeutic efficacy. This nanoplatform assembled by the outer-frame-degradable nanovehicles featuring NIR dual luminescence not only advances our understanding of where, when, and how therapeutic proteins take effect in vivo but also provides a universal route for visualizing the translocation of other bioactive macromolecules in cancer treatment and intervention.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Sustancias Luminiscentes , Nanoestructuras/química , Neoplasias/metabolismo , Proteínas Recombinantes , Animales , Femenino , Células HeLa , Humanos , Ácido Hialurónico/química , Rayos Infrarrojos , Sustancias Luminiscentes/análisis , Sustancias Luminiscentes/química , Sustancias Luminiscentes/farmacocinética , Ratones , Ratones Desnudos , Células 3T3 NIH , Proteínas Recombinantes/análisis , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Imagen Individual de Molécula/métodos
7.
Angew Chem Int Ed Engl ; 58(13): 4112-4128, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30182529

RESUMEN

With more and more engineered nanoparticles (NPs) being translated to the clinic, the United States Food and Drug Administration (FDA) has recently issued the latest draft guidance on nanomaterial-containing drug products with an emphasis on understanding their in vivo transport and nano-bio interactions. Following these guidelines, NPs can be designed to target and treat diseases more efficiently than small molecules, have minimum accumulation in normal tissues, and induce minimum toxicity. In this Minireview, we integrate this guidance with our ten-year studies on developing renal clearable luminescent gold NPs. These gold NPs resist serum protein adsorption, escape liver uptake, target cancerous tissues, and report kidney dysfunction at early stages. At the same time, off-target gold NPs can be eliminated by the kidneys with minimum accumulation in the body. Additionally, we identify challenges to the translation of renal clearable gold NPs from the bench to the clinic.


Asunto(s)
Oro/química , Riñón/metabolismo , Sustancias Luminiscentes/farmacocinética , Nanopartículas del Metal/química , Animales , Humanos , Luminiscencia , Sustancias Luminiscentes/química , Tasa de Depuración Metabólica , Nanopartículas del Metal/administración & dosificación , Distribución Tisular
8.
Am J Physiol Gastrointest Liver Physiol ; 315(4): G529-G537, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29927324

RESUMEN

Bile acid transporters, including the ileal apical sodium-dependent bile acid transporter (ASBT) and the hepatic sodium-taurocholate cotransporting polypeptide (NTCP), are crucial for the enterohepatic circulation of bile acids. Our objective was to develop a method for measuring bile acid transporter activity in real time to precisely evaluate rapid changes in their function. We designed a reporter system relying on a novel probe: cholic acid attached to luciferin via a disulfide-containing, self-immolating linker (CA-SS-Luc). Incubation of human embryonic kidney-293 cells coexpressing luciferase and ASBT with different concentrations of CA-SS-Luc (0.01-1 µM) resulted in bioluminescence with an intensity that was concentration- and time-dependent. The bioluminescence measured during incubation with 1 µM CA-SS-Luc was dependent on the levels of ASBT or NTCP expressed in the cells. Coincubation of CA-SS-Luc with natural bile acids enhanced the bioluminescence in a concentration-dependent manner with kinetic parameters for ASBT similar to those previously reported using conventional methods. These findings suggest that this method faithfully assesses ASBT function. Further, incubation with tyrosine phosphatase inhibitor III (PTPIII) led to significantly increased bioluminescence in cells expressing ASBT, consistent with previous studies showing an increase in ASBT function by PTPIII. We then investigated CA-SS-Luc in isolated mouse intestinal epithelial cells. Ileal enterocytes displayed significantly higher luminescence compared with jejunal enterocytes, indicating a transport process mediated by ileal ASBT. In conclusion, we have developed a novel method to monitor the activity of bile acid transporters in real time that has potential applications both for in vitro and in vivo studies. NEW & NOTEWORTHY This article reports the development of a real-time method for measuring the uptake of bile acids using a bioluminescent bile acid-based probe. This method has been validated for measuring uptake via the apical sodium-dependent bile acid transporter and the sodium-taurocholate cotransporting polypeptide in cell culture and ex vivo intestinal models.


Asunto(s)
Enterocitos/metabolismo , Luciferina de Luciérnaga/química , Sustancias Luminiscentes/química , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Simportadores/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Transporte Biológico Activo , Células Cultivadas , Ácido Cólico/química , Disulfuros/química , Femenino , Luciferina de Luciérnaga/farmacocinética , Células HEK293 , Humanos , Sustancias Luminiscentes/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente/métodos
9.
Inorg Chem ; 56(24): 15159-15170, 2017 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-29172469

RESUMEN

Hetero-bimetallic and -trimetallic complexes were synthesized by the combination of different metallic fragments, a luminescent Re(I) species, and a bioactive Au(I) derivative. A ditopic P,N-donor ligand (L) was used as linker between both metals, affording six new bipyridine (bipy) Re(I)/Au(I) hetero-metallic complexes of the type fac-[Re(bipy)(CO)3(LAuCl)]+ (4-6) and [(fac-[Re(bipy)(CO)3(L)])2Au]3+ (7-9) after a thorough synthetic procedure. Their emission is associated with a triplet metal-to-ligand charge transfer (Re(dπ) → bipy(π*)) transition and red-shifted in polar solvents with lifetimes in the range of nanoseconds and quantum yield values up to 12.5%. Cytotoxicity values in A549 cells of hetero-trimetallic species are almost twice that for the hetero-bimetallic (ca. 37 vs 69 µM, respectively), being the L-Au fragment the source of the antiproliferative activity. Species 7 and 8 showed similar behavior by fluorescence microscopy, with a nonuniform cytoplasmatic distribution, a clear accumulation in single spots at the edge of the inner cell membrane as well as in areas within the nucleus. Preliminary studies suggest the DNA as one of the targets and passive diffusion as the entrance pathway.


Asunto(s)
2,2'-Dipiridil/química , Antineoplásicos/química , Complejos de Coordinación/química , Oro/química , Sustancias Luminiscentes/química , Renio/química , 2,2'-Dipiridil/farmacocinética , 2,2'-Dipiridil/farmacología , Células A549 , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/farmacocinética , Complejos de Coordinación/farmacología , Oro/farmacocinética , Oro/farmacología , Humanos , Ligandos , Luminiscencia , Sustancias Luminiscentes/farmacocinética , Sustancias Luminiscentes/farmacología , Neoplasias/tratamiento farmacológico , Imagen Óptica , Renio/farmacocinética , Renio/farmacología
10.
Biomater Sci ; 5(8): 1393-1406, 2017 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-28484751

RESUMEN

Renal-clearable inorganic nanoparticles (NPs) that hold great potential in the future clinic translations are considered as the next generation of nanomedicine. In the past decade, enormous efforts have been dedicated to the development of renal-clearable NPs with fascinating optical properties, selective disease-targeting capabilities and low nanotoxicities. A further understanding of the design of renal-clearable luminescent metal NPs and their metabolic behavior in the body is important to achieve their clinical transition and extend their bioapplications in disease theranostics. In this review, we discuss the recent synthetic strategies of renal-clearable metal NPs in terms of the considerations of size and composition, surface chemistry and emission wavelength. We also summarize the current disease-related applications of these renal-clearable luminescent metal NPs in tumor targeting, kidney disease and antimicrobial investigations after a discussion of their biological behavior including the pharmacokinetics and biodistribution. Finally, we provide perspectives on the current challenges and upcoming chances for renal-clearable luminescent metal NPs.


Asunto(s)
Riñón/metabolismo , Sustancias Luminiscentes/química , Sustancias Luminiscentes/metabolismo , Nanopartículas del Metal , Animales , Humanos , Sustancias Luminiscentes/farmacocinética , Distribución Tisular
11.
Adv Healthc Mater ; 6(16)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28504415

RESUMEN

Gold nanoclusters are emerging as new materials for biomedical applications because of promises offered by their ultrasmall size and excellent biocompatibility. Here, the synthesis and optical and biological characterizations of a highly luminescent folate-functionalized Au22 cluster (Au22 -FA) are reported. The Au22 -FA clusters are synthesized by functionalizing the surface of Au22 (SG)18 clusters, where SG is glutathione, with benzyl chloroformate and folate. The functionalized clusters are highly water-soluble and exhibit remarkably bright luminescence with a quantum yield of 42%, significantly higher than any other water-soluble gold clusters protected with thiolate ligands. The folate groups conjugated to the gold cluster give rise to additional luminescence enhancement by energy transfer sensitization. The brightness of Au22 -FA is found to be 4.77 mM-1 cm-1 , nearly 8-fold brighter than that of Au22 (SG)18 . Further biological characterizations have revealed that the Au22 -FA clusters are well-suited for bioimaging. The Au22 -FA clusters exhibit excellent photostability and low toxicity; nearly 80% cell viability at 1000 ppm of the cluster. Additionally, the Au22 -FA clusters show target specificity to folate-receptor positive cells. Finally, the time-course in vivo luminescence images of intravenous-injected mice show that the Au22 -FA clusters are renal-clearable, leaving only 8% of them remained in the body after 24 h post-injection.


Asunto(s)
Ácido Fólico/química , Oro/química , Sustancias Luminiscentes/química , Nanopartículas del Metal/química , Imagen Molecular/métodos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ácido Fólico/farmacocinética , Glutatión/química , Glutatión/farmacocinética , Glutatión/toxicidad , Oro/farmacocinética , Humanos , Sustancias Luminiscentes/farmacocinética , Sustancias Luminiscentes/toxicidad , Nanopartículas del Metal/toxicidad , Ratones , Nanomedicina
12.
Ann Plast Surg ; 78(2): 217-222, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27845964

RESUMEN

Plastic and reconstructive surgeons increasingly apply adipose tissue grafting in a clinical setting, although the anticipation of graft survival is insecure. There are only few tools for tracking transplanted fat grafts in vivo.Murine adipose tissue clusters were incubated with negatively charged, mercaptoproprionic acid-coated cadmium telluride quantum dots (QDs) emitting in the dark red or near infrared. The intracellular localization of QDs was studied by confocal laser scanning microscopy.As a result, the adipose tissue clusters showed a proportional increase in fluorescence with increasing concentrations (1, 10, 16, 30, 50 nM) of cadmium telluride QDs. Laser scanning microscopy demonstrated a membrane bound localization of QDs. Vacuoles and cell nuclei of adipocytes were spared by QDs. We conclude that QDs were for the first time proven intracellular in adult adipocytes and demonstrate a strong fluorescence signal. Therefore, they may play an essential role for in vivo tracking of fat grafts.


Asunto(s)
Compuestos de Cadmio , Sustancias Luminiscentes , Puntos Cuánticos , Grasa Subcutánea/diagnóstico por imagen , Grasa Subcutánea/trasplante , Telurio , Animales , Compuestos de Cadmio/administración & dosificación , Compuestos de Cadmio/farmacocinética , Sustancias Luminiscentes/administración & dosificación , Sustancias Luminiscentes/farmacocinética , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Puntos Cuánticos/administración & dosificación , Espectroscopía Infrarroja Corta , Grasa Subcutánea/metabolismo , Telurio/administración & dosificación , Telurio/farmacocinética
13.
Sci Rep ; 6: 20418, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26853103

RESUMEN

Physiologic barriers such as the blood placenta barrier (BPB) and the blood brain barrier protect the underlying parenchyma from pathogens and toxins. ATP-binding cassette (ABC) transporters are transmembrane proteins found at these barriers, and function to efflux xenobiotics and maintain chemical homeostasis. Despite the plethora of ex vivo and in vitro data showing the function and expression of ABC transporters, no imaging modality exists to study ABC transporter activity in vivo at the BPB. In the present study, we show that in vitro models of the placenta possess ABCG2 activity and can specifically transport D-luciferin, the endogenous substrate of firefly luciferase. To test ABCG2 transport activity at the BPB, we devised a breeding strategy to generate a bioluminescent pregnant mouse model to demonstrate transporter function in vivo. We found that coadministering the ABCG2 inhibitors Ko143 and gefitinib with D-luciferin increased bioluminescent signal from fetuses and placentae, whereas the control P-gp inhibitor DCPQ had no effect. We believe that our bioluminescent pregnant mouse model will facilitate greater understanding of the BPB and ABCG2 activity in health and disease.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Transporte Biológico/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Coriocarcinoma/metabolismo , Procesamiento de Imagen Asistido por Computador/métodos , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Benzotiazoles/farmacocinética , Benzotiazoles/farmacología , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/efectos de los fármacos , Western Blotting , Células Cultivadas , Coriocarcinoma/tratamiento farmacológico , Coriocarcinoma/patología , Femenino , Citometría de Flujo , Gefitinib , Luciferasas de Luciérnaga/farmacocinética , Sustancias Luminiscentes/farmacocinética , Mediciones Luminiscentes , Masculino , Ratones , Ratones Transgénicos , Placenta , Embarazo , Quinazolinas/farmacocinética , Quinazolinas/farmacología , Distribución Tisular
14.
J Vis Exp ; (107): e53287, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26863490

RESUMEN

In contrast to commonly reported human glioma xenograft animal models, GL261 murine glioma xenografts recapitulate nearly all relevant clinical and histopathologic features of the human disease. When GL261 cells are implanted intracranially in syngeneic C57BL/6 mice, the model has the added advantage of maintaining an intact immune microenvironment. Stable expression of luciferase in GL261 cells allows non-invasive cost effective bioluminescence monitoring of intracranial tumor growth. We have recently demonstrated that luciferase expression in GL261 cells does not affect the tumor growth properties, tumor cell immunomodulatory cytokine expression, infiltration of immune cells into the tumor, or overall survival of animals bearing the intracranial tumor. Therefore, it appears that the GL261 luciferase glioma model can be useful in the study of novel chemotherapeutic and immunotherapeutic modalities. Here we report the technique for generating stable luciferase expression in GL261 cells and how to study the in vitro and in vivo growth of the tumor cells by bioluminescence imaging.


Asunto(s)
Transferencia de Energía por Resonancia de Bioluminiscencia/métodos , Neoplasias Encefálicas/diagnóstico por imagen , Glioblastoma/diagnóstico por imagen , Huésped Inmunocomprometido , Neoplasias Experimentales , Animales , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/inmunología , Línea Celular Tumoral , Glioblastoma/enzimología , Glioblastoma/inmunología , Luciferasas/farmacocinética , Sustancias Luminiscentes/farmacocinética , Ratones , Ratones Endogámicos C57BL
15.
Photodiagnosis Photodyn Ther ; 13: 244-254, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26253653

RESUMEN

Afterglow nanoparticles have been widely investigated as new agents for cancer imaging and as a light source for photodynamic activation for cancer treatment. For both applications, the targeting of the afterglow nanoparticles to tumor cells is an important and challenging issue. Here we report the strategies for targeting Sr3MgSi2O8:Eu(2+),Dy(3+) afterglow nanoparticles to tumor cells by conjugating with variety of targeting molecules such as folic acid, RGD peptide, and R-11 peptide. For folic acid targeting, experimental observations were conducted on PC-3 cells (folate receptor negative), MCF-7 (folate receptor positive), and KB cells (folate receptor positive) to compare the cellular uptake and confirm targeted delivery. For the cyclic RGDfK peptide, experiments were carried out on the integrin αvß3 positive MDA-MB-231 breast cancer cell line and the integrin αvß3 negative MCF-7 breast cancer cell lines in order to compare the cellular uptakes. As for R11-SH peptide, cellular uptake of the afterglow nanoparticles was observed on LNCaP and PC3 prostate cancer cell lines. All the observations showed that the cellular uptakes of the nanoparticles were enhanced by conjugation to variety of targeting molecules which are specific for breast and prostate cancer cells.


Asunto(s)
Sustancias Luminiscentes/síntesis química , Sustancias Luminiscentes/farmacocinética , Nanoconjugados/química , Nanopartículas/química , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Línea Celular Tumoral , Humanos , Ensayo de Materiales , Nanoconjugados/ultraestructura , Nanopartículas/ultraestructura
17.
J Nanosci Nanotechnol ; 15(4): 2798-803, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26353495

RESUMEN

In this study, four kinds of upconversion nanocrystals (UCNs) have been successfully synthesized by a facile solvothermal method. The morphology, crystalline phase, composition, grain size, upconversion luminescence and cell image of the UCNs were investigated. The properties of the NaLuF4-based UCNs were compared with the counterparts of NaYF4-based UCNs. It is found that the NaLuF4-based UCNs are apt to form hexagonal phase structures, while NaYF4-based UCNs of NaYF4:Yb, Er and NaYF4:Gd, Yb, Er are cubic and hexagonal phases respectively. The upconversion emission intensities of the NaLuF4-based UCNs are higher than that of NaYF4-based UCNs, and Gd3+ presented UCNs are higher than that of Gd3+ absented UCNs. The bioimaging application of NaLuF4:Gd, Yb, Er shows that bright upconversion luminescence can be observed when UCNs-labeled HeLa cells are excited with 980 nm light.


Asunto(s)
Fluoruros/química , Lutecio/química , Nanopartículas/química , Nanotecnología/métodos , Itrio/química , Membrana Celular/química , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Fluoruros/farmacocinética , Fluoruros/toxicidad , Células HeLa , Humanos , Sustancias Luminiscentes/química , Sustancias Luminiscentes/farmacocinética , Sustancias Luminiscentes/toxicidad , Lutecio/farmacocinética , Lutecio/toxicidad , Nanopartículas/toxicidad , Itrio/farmacocinética , Itrio/toxicidad
18.
J Nanobiotechnology ; 13: 38, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26040273

RESUMEN

BACKGROUND: The fertility performance of animals is still a mystery and the full comprehension of mammalian gametes maturation and early embryonic development remains to be elucidated. The recent development in nanotechnology offers a new opportunity for real-time study of reproductive cells in their physiological environments. As a first step toward that goal, we evaluated the effectiveness of a fluorescent and luminescent nanoparticle for in vitro and ex vivo imaging of porcine gametes. METHODS: Freshly harvested boar sperm were labeled with red-shifted (655 nm) quantum dot nanoparticles conjugated (QD+) or not (QD-) with plasminogen antibody and evaluated. Subsets of labeled spermatozoa were loaded into straws and placed within the lumen of gilt reproductive tracts for ex vivo intra-uterine imaging. Porcine cumulus-oocyte complexes (COCs) were matured in the presence of QD- or QD+. Ovarian follicles were microinjected with QD- or QD+ and placed in culture for up to 4 days. After labeling, all samples were supplemented with coelenterazine, the luciferase substrate, and immediately submitted to bioluminescence analysis, followed by fluorescence and hyperspectral imaging. Data were analyzed with ANOVA and P < 0.05 indicated significant differences. RESULTS: All labeled-samples revealed bioluminescence emission that was confirmed by fluorescence and hyperspectral imaging of the QD localization within the cells and tissues. Over 76% of spermatozoa and both immature and mature COCs were successfully labeled with QD- or QD+. The QD- fluorescence appeared homogenously distributed in the oocytes, while found in the entire sperm length with a higher accumulation within the mid-piece. Labeled-follicles exhibited a progressive migration of QD nanoparticles within the follicle wall during culture. In contrast, QD+ fluorescence signals appeared condensed and stronger in the follicle cells, sperm head, and sub-plasma membrane area of mature oocytes. Weaker QD+ signals were detected in the cumulus cells. Fluorescence and hyperspectral microscope imaging showed comparable intracellular QD localization. Ex-vivo intra-uterine bioluminescence imaging of labeled spermatozoa revealed stronger signals captured over the oviducts, with uterine body allowing the lowest signal detection. CONCLUSION: Findings indicate that conjugated and non-conjugated fluorescent nanoparticles can be used for effective labeling of mammalian gametes for in vitro monitoring and potential in vivo targeted-imaging.


Asunto(s)
Sustancias Luminiscentes/farmacocinética , Mediciones Luminiscentes/métodos , Oocitos/fisiología , Puntos Cuánticos , Espermatozoides/fisiología , Animales , Supervivencia Celular , Femenino , Genitales Femeninos/fisiología , Luciferasas de Renilla/química , Sustancias Luminiscentes/química , Mediciones Luminiscentes/instrumentación , Masculino , Microscopía Confocal/métodos , Microscopía Electrónica de Transmisión , Nanopartículas/química , Oocitos/química , Folículo Ovárico/fisiología , Espermatozoides/química , Porcinos
19.
Nanotechnology ; 26(21): 215703, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25943071

RESUMEN

Designing various inorganic nanomaterials that are cost effective, water soluble, optically photostable, highly fluorescent and biocompatible for bioimaging applications is a challenging task. Similar to semiconducting quantum dots (QDs), silicon QDs are another alternative and are highly fluorescent, but non-water soluble. Several surface modification strategies were adopted to make them water soluble. However, the photoluminescence of Si QDs was seriously quenched in the aqueous environment. In this report, highly luminescent, water-dispersible, blue- and green-emitting Si QDs were prepared with good photostability. In vitro studies in monocytes reveal that Si QDs exhibit good biocompatibility and excellent distribution throughout the cytoplasm region, along with the significant fraction translocated into the nucleus. The in vivo zebrafish studies also reveal that Si QDs can be evenly distributed in the yolk-sac region. Overall, our results demonstrate the applicability of water-soluble and highly fluorescent Si QDs as excellent in vitro and in vivo bioimaging probes.


Asunto(s)
Materiales Biocompatibles/química , Materiales Biocompatibles/toxicidad , Medios de Contraste/química , Medios de Contraste/toxicidad , Puntos Cuánticos/química , Puntos Cuánticos/toxicidad , Animales , Materiales Biocompatibles/farmacocinética , Células Cultivadas , Medios de Contraste/farmacocinética , Sustancias Luminiscentes/química , Sustancias Luminiscentes/farmacocinética , Sustancias Luminiscentes/toxicidad , Mediciones Luminiscentes , Ensayo de Materiales , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Silicio/química , Silicio/farmacocinética , Solubilidad , Agua , Pez Cebra
20.
Mol Imaging ; 132014.
Artículo en Inglés | MEDLINE | ID: mdl-25249435

RESUMEN

Stem cell therapy has been studied intensively as a promising therapeutic strategy toward a cure for diabetes. To study the effect of mesenchymal stem cell (MSC) transplantation for pancreatic regeneration, we monitored the localization and distribution of transplanted MSCs by bioluminescence imaging in a mouse model. Bone marrow MSCs were isolated and transfected with a highly sensitive firefly luciferase reporter gene. To assess the efficiency of MSC transplantation, a partially pancreatectomized (PPx) mouse model was used. Transplanted MSCs were monitored by confocal microscopy and in vivo bioluminescence imaging. Daily blood glucose levels and glucose tolerance were measured. Insulin-secreting beta cells were immunostained, and insulin levels were measured via enzyme-linked immunosorbent assay. Bioluminescence signals were clearly detected from the transplanted MSCs in the pancreatic region regardless of injection route. However, locally injected MSCs exhibited more rapid proliferation than ductally injected MSCs. PPx mice harboring transplanted MSCs gradually recovered from impaired glucose tolerance. Although insulin secretion was not observed in MSCs, transplanted MSCs facilitate the injured pancreas to recover its function. In vivo optical imaging of transplanted MSCs using a highly sensitive luciferase reporter enables the assessment of MSC transplantation efficiency in a PPx mouse model.


Asunto(s)
Células de la Médula Ósea/citología , Sustancias Luminiscentes/farmacocinética , Células Madre Mesenquimatosas/citología , Páncreas/patología , Animales , Células Cultivadas , Técnicas de Cocultivo , Femenino , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Células HEK293 , Humanos , Luciferasas de Luciérnaga/farmacocinética , Mediciones Luminiscentes , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Modelos Animales , Páncreas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...