Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 2: 326, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31508501

RESUMO

The kidney's inherent complexity has made identifying cell-specific pathways challenging, particularly when temporally associating them with the dynamic pathophysiology of acute kidney injury (AKI). Here, we combine renal cell-specific luciferase reporter mice using a chemoselective luciferin to guide the acquisition of cell-specific transcriptional changes in C57BL/6 background mice. Hydrogen peroxide generation, a common mechanism of tissue damage, was tracked using a peroxy-caged-luciferin to identify optimum time points for immunoprecipitation of labeled ribosomes for RNA-sequencing. Together, these tools revealed a profound impact of AKI on mitochondrial pathways in the collecting duct. In fact, targeting the mitochondria with an antioxidant, ameliorated not only hydrogen peroxide generation, but also significantly reduced oxidative stress and the expression of the AKI biomarker, LCN2. This integrative approach of coupling physiological imaging with transcriptomics and drug testing revealed how the collecting duct responds to AKI and opens new venues for cell-specific predictive monitoring and treatment.


Assuntos
Injúria Renal Aguda/genética , Imageamento Tridimensional , Isquemia/genética , Isquemia/patologia , Transcriptoma/genética , Injúria Renal Aguda/complicações , Injúria Renal Aguda/patologia , Animais , Antioxidantes/metabolismo , Túbulos Renais Coletores/lesões , Túbulos Renais Coletores/patologia , Camundongos Endogâmicos C57BL , Néfrons/metabolismo , Néfrons/patologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
2.
Cell Metab ; 27(3): 689-701.e4, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29514074

RESUMO

Metabolism is a fundamental process of life. However, non-invasive measurement of local tissue metabolism is limited today by a deficiency in adequate tools for in vivo observations. We designed a multi-modular platform that explored the relation between local tissue oxygen consumption, determined by label-free optoacoustic measurements of hemoglobin, and concurrent indirect calorimetry obtained during metabolic activation of brown adipose tissue (BAT). By studying mice and humans, we show how video-rate handheld multi-spectral optoacoustic tomography (MSOT) in the 700-970 nm spectral range enables non-invasive imaging of BAT activation, consistent with positron emission tomography findings. Moreover, we observe BAT composition differences between healthy and diabetic tissues. The study consolidates hemoglobin as a principal label-free biomarker for longitudinal non-invasive imaging of BAT morphology and bioenergetics in situ. We also resolve water and fat components in volunteers, and contrast MSOT readouts with magnetic resonance imaging data.


Assuntos
Tecido Adiposo Marrom/metabolismo , Hemoglobinas/metabolismo , Consumo de Oxigênio , Técnicas Fotoacústicas , Adulto , Animais , Metabolismo Energético , Feminino , Humanos , Metabolismo dos Lipídeos , Masculino , Camundongos Endogâmicos BALB C
3.
Head Neck ; 39(1): 98-108, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27437903

RESUMO

BACKGROUND: The correlation between podoplanin expression and extracapsular spread in head and neck squamous cell carcinoma (HNSCC) has never been reported. The purpose of this study was to assess the predictive value of podoplanin expression for this parameter. METHODS: Subjective immunoreactivity scores and semiquantitative image analysis of podoplanin expression were performed in 67 patients with primary oral squamous cell carcinoma and in their corresponding lymph nodes. Neck classification showed 34 cases (51%) of pN0 and 33 cases (49%) of pN+. Correlation between the levels of podoplanin expression and the histopathological data was established. RESULTS: In lymph nodes, a high level of podoplanin expression correlated with the presence of extracapsular spread by multivariate analysis (p = .03). A strong correlation between subjective and semiquantitative image analysis was observed (r = 0.77; p < .001). CONCLUSION: A high level of podoplanin expression in lymph node metastases of oral squamous cell carcinoma is independently associated with extracapsular spread. © 2016 Wiley Periodicals, Head Neck 39: 98-108, 2017.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Glicoproteínas de Membrana/metabolismo , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Estudos Retrospectivos
4.
Gastroenterology ; 152(1): 78-81.e2, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27742378

RESUMO

Alterations in hepatic free fatty acid (FFA) uptake and metabolism contribute to the development of prevalent liver disorders such as hepatosteatosis. However, detecting dynamic changes in FFA uptake by the liver in live model organisms has proven difficult. To enable noninvasive real-time imaging of FFA flux in the liver, we generated transgenic mice with liver-specific expression of luciferase and performed bioluminescence imaging with an FFA probe. Our approach enabled us to observe the changes in FFA hepatic uptake under different physiological conditions in live animals. By using this method, we detected a decrease in FFA accumulation in the liver after mice were given injections of deoxycholic acid and an increase after they were fed fenofibrate. In addition, we observed diurnal regulation of FFA hepatic uptake in living mice. Our imaging system appears to be a useful and reliable tool for studying the dynamic changes in hepatic FFA flux in models of liver disease.


Assuntos
Ácidos Graxos/metabolismo , Fígado/diagnóstico por imagem , Fígado/metabolismo , Medições Luminescentes , Animais , Transporte Biológico/efeitos dos fármacos , Colagogos e Coleréticos/farmacologia , Ritmo Circadiano , Ácido Desoxicólico/farmacologia , Fenofibrato/farmacologia , Hipolipemiantes/farmacologia , Luciferases/genética , Masculino , Camundongos , Camundongos Transgênicos , Fotografação
5.
J Am Acad Orthop Surg ; 25 Suppl 1: S7-S12, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27941556

RESUMO

INTRODUCTION: Diagnosing prosthetic joint infection (PJI) poses significant challenges, and current modalities are fraught with low sensitivity and/or potential morbidity. Photoacoustic imaging (PAI) is a novel ultrasound-based modality with potential for diagnosing PJI safely and noninvasively. MATERIALS: In an established preclinical mouse model of bioluminescent Staphylococcus aureus PJI, fluorescent indocyanine green (ICG) was conjugated to ß-cyclodextrin (CDX-ICG) or teicoplanin (Teic-ICG) and injected intravenously for 1 week postoperatively. Daily fluorescent imaging and PAI were used to localize and quantify tracer signals. Results were analyzed using 2-way analysis of variance. RESULTS: Fluorescence clearly localized to the site of infection and was significantly higher with Teic-ICG compared with CDX-ICG (P = 0.046) and ICG alone (P = 0.0087). With PAI, the photoacoustic signal per volumetric analysis was substantially higher and better visualized with Teic-ICG compared with CDX-ICG and ICG alone, and colocalized well with bioluminescence and fluorescence imaging. CONCLUSION: Photoacoustic imaging successfully localized PJI in this proof-of-concept study and demonstrates potential for clinical translation in orthopaedics.


Assuntos
Artroplastia de Substituição/efeitos adversos , Técnicas Fotoacústicas/métodos , Infecções Relacionadas à Prótese/diagnóstico por imagem , Animais , Medições Luminescentes/métodos , Masculino , Camundongos Endogâmicos C57BL , Imagem Óptica/métodos
6.
Front Oncol ; 6: 221, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27818949

RESUMO

PURPOSE: Most effective antitumor therapies induce tumor cell death. Non-invasive, rapid and accurate quantitative imaging of cell death is essential for monitoring early response to antitumor therapies. To facilitate this, we previously developed a biocompatible necrosis-avid near-infrared fluorescence (NIRF) imaging probe, HQ4, which was radiolabeled with 111Indium-chloride (111In-Cl3) via the chelate diethylene triamine pentaacetic acid (DTPA), to enable clinical translation. The aim of the present study was to evaluate the application of HQ4-DTPA for monitoring tumor cell death induced by radiation therapy. Apart from its NIRF and radioactive properties, HQ4-DTPA was also tested as a photoacoustic imaging probe to evaluate its performance as a multimodal contrast agent for superficial and deep tissue imaging. MATERIALS AND METHODS: Radiation-induced tumor cell death was examined in a xenograft mouse model of human breast cancer (MCF-7). Tumors were irradiated with three fractions of 9 Gy each. HQ4-DTPA was injected intravenously after the last irradiation, NIRF and photoacoustic imaging of the tumors were performed at 12, 20, and 40 h after injection. Changes in probe accumulation in the tumors were measured in vivo, and ex vivo histological analysis of excised tumors was performed at experimental endpoints. In addition, biodistribution of radiolabeled [111In]DTPA-HQ4 was assessed using hybrid single-photon emission computed tomography-computed tomography (SPECT-CT) at the same time points. RESULTS: In vivo NIRF imaging demonstrated a significant difference in probe accumulation between control and irradiated tumors at all time points after injection. A similar trend was observed using in vivo photoacoustic imaging, which was validated by ex vivo tissue fluorescence and photoacoustic imaging. Serial quantitative radioactivity measurements of probe biodistribution further demonstrated increased probe accumulation in irradiated tumors. CONCLUSION: HQ4-DTPA has high specificity for dead cells in vivo, potentiating its use as a contrast agent for determining the relative level of tumor cell death following radiation therapy using NIRF, photoacoustic imaging and SPECT in vivo. Initial preclinical results are promising and indicate the need for further evaluation in larger cohorts. If successful, such studies may help develop a new multimodal method for non-invasive and dynamic deep tissue imaging of treatment-induced cell death to quantitatively assess therapeutic response in patients.

7.
Mol Imaging Biol ; 18(6): 905-915, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27277828

RESUMO

PURPOSE: Recently we showed that a number of carboxylated near-infrared fluorescent (NIRF) cyanine dyes possess strong necrosis avid properties in vitro as well as in different mouse models of spontaneous and therapy-induced tumor necrosis, indicating their potential use for cancer diagnostic- and prognostic purposes. In the previous study, the detection of the cyanines was achieved by whole body optical imaging, a technique that, due to the limited penetration of near-infrared light, is not suitable for investigations deeper than 1 cm within the human body. Therefore, in order to facilitate clinical translation, the purpose of the present study was to generate a necrosis avid cyanine-based NIRF probe that could also be used for single photon emission computed tomography (SPECT). For this, the necrosis avid NIRF cyanine HQ4 was radiolabeled with 111indium, via the chelate diethylene triamine pentaacetic acid (DTPA). PROCEDURES: The necrosis avid properties of the radiotracer [111In]DTPA-HQ4 were examined in vitro and in vivo in different breast tumor models in mice using SPECT and optical imaging. Moreover, biodistribution studies were performed to examine the pharmacokinetics of the probe in vivo. RESULTS: Using optical imaging and radioactivity measurements, in vitro, we showed selective accumulation of [111In]DTPA-HQ4 in dead cells. Using SPECT and in biodistribution studies, the necrosis avidity of the radiotracer was confirmed in a 4T1 mouse breast cancer model of spontaneous tumor necrosis and in a MCF-7 human breast cancer model of chemotherapy-induced tumor necrosis. CONCLUSIONS: The radiotracer [111In]DTPA-HQ4 possessed strong and selective necrosis avidity in vitro and in various mouse models of tumor necrosis in vivo, indicating its potential to be clinically applied for diagnostic purposes and to monitor anti-cancer treatment efficacy.


Assuntos
Carbocianinas/química , Imagem Multimodal/métodos , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Radioisótopos de Índio/química , Camundongos Endogâmicos BALB C , Camundongos Nus , Necrose , Imagem Óptica , Ácido Pentético/química , Distribuição Tecidual
8.
Head Neck ; 38(9): 1407-15, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27043718

RESUMO

BACKGROUND: The use of lymphatic vessel density as a predictor of occult lymph node metastasis (OLNM) in head and neck squamous cell carcinoma (HNSCC) has never been reported. METHODS: Staining of the specific lymphatic endothelial cells nuclear marker, PROX1, as an indicator of lymphatic vessel density was determined by counting the number of positive cells in squamous cell carcinomas (SCCs) of the oral cavity and the oropharynx with clinically negative necks. Correlation with histopathological data was established. RESULTS: Peritumoral PROX1 lymphatic nuclear count significantly correlated with the detection of OLNM in multivariate analysis (p < .005). The sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of this parameter was 60%, 95%, 85%, and 90%, respectively. CONCLUSION: Peritumoral PROX1 lymphatic nuclear count in primary SCCs of the oral cavity and the oropharynx allows accurate prediction of occult lymph node metastasis. © 2016 Wiley Periodicals, Inc. Head Neck 38: 1407-1415, 2016.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/patologia , Neoplasias de Cabeça e Pescoço/patologia , Proteínas de Homeodomínio/metabolismo , Linfonodos/patologia , Vasos Linfáticos/patologia , Neoplasias Bucais/patologia , Proteínas Supressoras de Tumor/metabolismo , Idoso , Análise de Variância , Estudos de Coortes , Feminino , Humanos , Imuno-Histoquímica , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , Boca/patologia , Análise Multivariada , Invasividade Neoplásica/patologia , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Prognóstico , Estudos Retrospectivos , Medição de Risco , Carcinoma de Células Escamosas de Cabeça e Pescoço , Suíça
9.
Diabetes ; 64(11): 3713-24, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26293504

RESUMO

Novel, clinically relevant, approaches to shift energy balance are urgently needed to combat metabolic disorders such as obesity and diabetes. One promising approach has been the expansion of brown adipose tissues that express uncoupling protein (UCP) 1 and thus can uncouple mitochondrial respiration from ATP synthesis. While expansion of UCP1-expressing adipose depots may be achieved in rodents via genetic and pharmacological manipulations or the transplantation of brown fat depots, these methods are difficult to use for human clinical intervention. We present a novel cell scaffold technology optimized to establish functional brown fat-like depots in vivo. We adapted the biophysical properties of hyaluronic acid-based hydrogels to support the differentiation of white adipose tissue-derived multipotent stem cells (ADMSCs) into lipid-accumulating, UCP1-expressing beige adipose tissue. Subcutaneous implantation of ADMSCs within optimized hydrogels resulted in the establishment of distinct UCP1-expressing implants that successfully attracted host vasculature and persisted for several weeks. Importantly, implant recipients demonstrated elevated core body temperature during cold challenges, enhanced respiration rates, improved glucose homeostasis, and reduced weight gain, demonstrating the therapeutic merit of this highly translatable approach. This novel approach is the first truly clinically translatable system to unlock the therapeutic potential of brown fat-like tissue expansion.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo Marrom/transplante , Canais Iônicos/metabolismo , Proteínas Mitocondriais/metabolismo , Células-Tronco/metabolismo , Termogênese/fisiologia , Alicerces Teciduais , Tecido Adiposo Marrom/metabolismo , Animais , Temperatura Corporal/fisiologia , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Temperatura Baixa , Metabolismo Energético/fisiologia , Camundongos , Proteína Desacopladora 1
10.
PLoS One ; 10(6): e0131037, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26110789

RESUMO

Bacterial nitroreductases (NTRs) have been widely utilized in the development of novel antibiotics, degradation of pollutants, and gene-directed enzyme prodrug therapy (GDEPT) of cancer that reached clinical trials. In case of GDEPT, since NTR is not naturally present in mammalian cells, the prodrug is activated selectively in NTR-transformed cancer cells, allowing high efficiency treatment of tumors. Currently, no bioluminescent probes exist for sensitive, non-invasive imaging of NTR expression. We therefore developed a "NTR caged luciferin" (NCL) probe that is selectively reduced by NTR, producing light proportional to the NTR activity. Here we report successful application of this probe for imaging of NTR in vitro, in bacteria and cancer cells, as well as in vivo in mouse models of bacterial infection and NTR-expressing tumor xenografts. This novel tool should significantly accelerate the development of cancer therapy approaches based on GDEPT and other fields where NTR expression is important.


Assuntos
Diagnóstico por Imagem/métodos , Medições Luminescentes/métodos , Neoplasias/diagnóstico , Nitrorredutases/metabolismo , Animais , Linhagem Celular , Feminino , Terapia Genética/métodos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Pró-Fármacos/farmacologia
11.
Nat Commun ; 5: 5830, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25518894

RESUMO

Super-resolution optical fluctuation imaging (SOFI) provides an elegant way of overcoming the diffraction limit in all three spatial dimensions by computing higher-order cumulants of image sequences of blinking fluorophores acquired with a classical widefield microscope. Previously, three-dimensional (3D) SOFI has been demonstrated by sequential imaging of multiple depth positions. Here we introduce a multiplexed imaging scheme for the simultaneous acquisition of multiple focal planes. Using 3D cross-cumulants, we show that the depth sampling can be increased. The simultaneous acquisition of multiple focal planes significantly reduces the acquisition time and thus the photobleaching. We demonstrate multiplane 3D SOFI by imaging fluorescently labelled cells over an imaged volume of up to 65 × 65 × 3.5 µm(3) without depth scanning. In particular, we image the 3D network of mitochondria in fixed C2C12 cells immunostained with Alexa 647 fluorophores and the 3D vimentin structure in living Hela cells expressing the fluorescent protein Dreiklang.


Assuntos
Imageamento Tridimensional/instrumentação , Mitocôndrias/ultraestrutura , Imagem Molecular/instrumentação , Mioblastos/ultraestrutura , Animais , Carbocianinas , Linhagem Celular , Corantes Fluorescentes , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Imageamento Tridimensional/métodos , Camundongos , Microscopia de Fluorescência/métodos , Mitocôndrias/fisiologia , Imagem Molecular/métodos , Mioblastos/fisiologia , Vimentina/química , Vimentina/metabolismo
12.
Curr Protoc Chem Biol ; 6(3): 169-189, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25205565

RESUMO

The great complexity of many human pathologies, such as cancer, diabetes, and neurodegenerative diseases, requires new tools for studies of biological processes on the whole organism level. The discovery of novel biocompatible reactions has tremendously advanced our understanding of basic biology; however, no efficient tools exist for real-time non-invasive imaging of many human proteases that play very important roles in multiple human disorders. We recently reported that the "split luciferin" biocompatible reaction represents a valuable tool for evaluation of protease activity directly in living animals using bioluminescence imaging (BLI). Since BLI is the most sensitive in vivo imaging modality known to date, this method can be widely applied for the evaluation of the activity of multiple proteases, as well as identification of their new peptide-specific substrates. In this unit, we describe several applications of this "split luciferin" reaction for quantification of protease activities in test tube assays and living animals.


Assuntos
Benzotiazóis/química , Corantes Fluorescentes/química , Imagem Molecular/métodos , Peptídeo Hidrolases/química , Animais , Animais Geneticamente Modificados , Caspase 3/química , Caspase 3/metabolismo , Caspase 7/química , Caspase 7/metabolismo , Modelos Animais de Doenças , Luminescência , Camundongos , Peptídeo Hidrolases/metabolismo , Trombina/química , Trombina/metabolismo
13.
Methods Enzymol ; 538: 107-34, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24529436

RESUMO

The ability of white and brown adipose tissue to efficiently take up long-chain fatty acids is key to their physiological functions in energy storage and thermogenesis, respectively. Several approaches have been taken to determine uptake rates by cultured cells and primary adipocytes including radio- and fluorescently labeled fatty acids. In addition, the recent description of activatable bioluminescent fatty acids has opened the possibility for expanding these in vitro approaches to real-time monitoring of fatty acid uptake kinetics by adipose depots in vivo. Here, we will describe some of the most useful experimental paradigms to quantitatively determine long-chain fatty acid uptake by adipocytes in vitro and provide the reader with detailed instruction on how bioluminescent probes for in vivo imaging can be synthesized and used in living mice.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Ácidos Graxos/análise , Ácidos Graxos/metabolismo , Medições Luminescentes/métodos , Imagem Óptica/métodos , Células 3T3-L1 , Tecido Adiposo/citologia , Animais , Transporte Biológico , Compostos de Boro/análise , Compostos de Boro/metabolismo , Células Cultivadas , Corantes Fluorescentes/análise , Corantes Fluorescentes/metabolismo , Humanos , Substâncias Luminescentes/análise , Substâncias Luminescentes/metabolismo , Camundongos , Sondas Moleculares/análise , Sondas Moleculares/metabolismo , Imagem Corporal Total/métodos
14.
ACS Chem Biol ; 8(5): 987-99, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23463944

RESUMO

The discovery of biocompatible reactions had a tremendous impact on chemical biology, allowing the study of numerous biological processes directly in complex systems. However, despite the fact that multiple biocompatible reactions have been developed in the past decade, very few work well in living mice. Here we report that D-cysteine and 2-cyanobenzothiazoles can selectively react with each other in vivo to generate a luciferin substrate for firefly luciferase. The success of this "split luciferin" ligation reaction has important implications for both in vivo imaging and biocompatible labeling strategies. First, the production of a luciferin substrate can be visualized in a live mouse by bioluminescence imaging (BLI) and furthermore allows interrogation of targeted tissues using a "caged" luciferin approach. We therefore applied this reaction to the real-time noninvasive imaging of apoptosis associated with caspase 3/7. Caspase-dependent release of free D-cysteine from the caspase 3/7 peptide substrate Asp-Glu-Val-Asp-D-Cys (DEVD-(D-Cys)) allowed selective reaction with 6-amino-2-cyanobenzothiazole (NH(2)-CBT) in vivo to form 6-amino-D-luciferin with subsequent light emission from luciferase. Importantly, this strategy was found to be superior to the commercially available DEVD-aminoluciferin substrate for imaging of caspase 3/7 activity. Moreover, the split luciferin approach enables the modular construction of bioluminogenic sensors, where either or both reaction partners could be caged to report on multiple biological events. Lastly, the luciferin ligation reaction is 3 orders of magnitude faster than Staudinger ligation, suggesting further applications for both bioluminescence and specific molecular targeting in vivo.


Assuntos
Benzotiazóis/química , Benzotiazóis/síntese química , Luciferases de Vaga-Lume/metabolismo , Substâncias Luminescentes/química , Medições Luminescentes/métodos , Nitrilas/química , Peptídeo Hidrolases/análise , Peptídeo Hidrolases/metabolismo , Animais , Apoptose , Benzotiazóis/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Cisteína/química , Feminino , Luciferina de Vaga-Lumes/análogos & derivados , Luciferina de Vaga-Lumes/metabolismo , Humanos , Cinética , Luciferases de Vaga-Lume/genética , Substâncias Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Oligopeptídeos/metabolismo , Trombina/metabolismo
15.
ACS Chem Biol ; 7(11): 1884-91, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22928772

RESUMO

Detection and quantification of fatty acid fluxes in animal model systems following physiological, pathological, or pharmacological challenges is key to our understanding of complex metabolic networks as these macronutrients also activate transcription factors and modulate signaling cascades including insulin sensitivity. To enable noninvasive, real-time, spatiotemporal quantitative imaging of fatty acid fluxes in animals, we created a bioactivatable molecular imaging probe based on long-chain fatty acids conjugated to a reporter molecule (luciferin). We show that this probe faithfully recapitulates cellular fatty acid uptake and can be used in animal systems as a valuable tool to localize and quantitate in real time lipid fluxes such as intestinal fatty acid absorption and brown adipose tissue activation. This imaging approach should further our understanding of basic metabolic processes and pathological alterations in multiple disease models.


Assuntos
Ácidos Graxos/análise , Ácidos Graxos/metabolismo , Imagem Molecular/métodos , Sondas Moleculares/análise , Células 3T3-L1 , Animais , Transporte Biológico , Expressão Gênica , Insulina/metabolismo , Luciferases/genética , Luciferases/metabolismo , Medições Luminescentes/métodos , Camundongos , Camundongos Transgênicos
16.
Proc Natl Acad Sci U S A ; 107(50): 21316-21, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21115844

RESUMO

Living organisms produce hydrogen peroxide (H(2)O(2)) to kill invading pathogens and for cellular signaling, but aberrant generation of this reactive oxygen species is a hallmark of oxidative stress and inflammation in aging, injury, and disease. The effects of H(2)O(2) on the overall health of living animals remain elusive, in part owing to a dearth of methods for studying this transient small molecule in vivo. Here we report the design, synthesis, and in vivo applications of Peroxy Caged Luciferin-1 (PCL-1), a chemoselective bioluminescent probe for the real-time detection of H(2)O(2) within living animals. PCL-1 is a boronic acid-caged firefly luciferin molecule that selectively reacts with H(2)O(2) to release firefly luciferin, which triggers a bioluminescent response in the presence of firefly luciferase. The high sensitivity and selectivity of PCL-1 for H(2)O(2), combined with the favorable properties of bioluminescence for in vivo imaging, afford a unique technology for real-time detection of basal levels of H(2)O(2) generated in healthy, living mice. Moreover, we demonstrate the efficacy of PCL-1 for monitoring physiological fluctuations in H(2)O(2) levels by directly imaging elevations in H(2)O(2) within testosterone-stimulated tumor xenografts in vivo. The ability to chemoselectively monitor H(2)O(2) fluxes in real time in living animals offers opportunities to dissect H(2)O(2)'s disparate contributions to health, aging, and disease.


Assuntos
Peróxido de Hidrogênio/metabolismo , Medições Luminescentes/métodos , Neoplasias Experimentais/metabolismo , Animais , Linhagem Celular Tumoral , Luciferina de Vaga-Lumes/metabolismo , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Estrutura Molecular , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Transplante Heterólogo
17.
J Am Chem Soc ; 132(25): 8563-5, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20527879

RESUMO

Cell-surface glycans are attractive targets for molecule imaging due to their reflection of cellular processes associated with development and disease progression. In this paper, we describe the design, synthesis, and biological application of a new phosphine probe for real-time imaging of cell-surface glycans using bioluminescence. To accomplish this goal, we took advantage of the bioorthogonal chemical reporter technique. This strategy uses a two-step labeling procedure in which an unnatural sugar analogue containing a functional handle is (1) incorporated into sugar-bearing proteins via the cell's own biosynthetic machinery and then (2) detected with an exogenously added probe. We designed phosphine-luciferin reagent 1 to activate bioluminescence in response to Staudinger ligation with azide-labeled glycans. We chose to use a phosphine probe because, despite their slow reaction kinetics, they remain the best-performing reagents for tagging azidosugars in mice. Given the sensitivity and negligible background provided by bioluminescence imaging (BLI), we reasoned that 1 might be able to overcome some of the limitations encountered with fluorescent phosphine probes. In this work, we synthesized the first phosphine-luciferin probe for use in real-time BLI and demonstrated that azide-labeled cell-surface glycans can be imaged with 1 using concentrations as low as single digit nanomolar and times as little as 5 min, a feat that cannot be matched by any previous fluorescent phosphine probes. Even though we have only demonstrated its use in visualizing glycans, it can be envisioned that this probe could also be used for bioluminescence imaging of any azide-containing biomolecule, such as proteins and lipids, since azides have been previously incorporated into these molecules. The phosphine-luciferin probe is therefore poised for many applications in real-time imaging in cells and whole animals. These studies are currently in progress in our laboratory.


Assuntos
Medições Luminescentes/métodos , Imagem Molecular/métodos , Polissacarídeos/metabolismo , Animais , Azidas/química , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Camundongos , Polissacarídeos/química , Fatores de Tempo
18.
Proc Natl Acad Sci U S A ; 105(34): 12128-33, 2008 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-18713866

RESUMO

Many cancer therapeutic agents elicit resistance that renders them ineffective and often produces cross-resistance to other drugs. One of the most common mechanisms of resistance involves P-glycoprotein (Pgp)-mediated drug efflux. To address this problem, new agents have been sought that are less prone to inducing resistance and less likely to serve as substrates for Pgp efflux. An alternative to this approach is to deliver established agents as molecular transporter conjugates into cells through a mechanism that circumvents Pgp-mediated efflux and allows for release of free drug only after cell entry. Here we report that the widely used chemotherapeutic agent Taxol, ineffective against Taxol-resistant human ovarian cancer cell lines, can be incorporated into a releasable octaarginine conjugate that is effective against the same Taxol-resistant cell lines. It is significant that the ability of the Taxol conjugates to overcome Taxol resistance is observed both in cell culture and in animal models of ovarian cancer. The generality and mechanistic basis for this effect were also explored with coelenterazine, a Pgp substrate. Although coelenterazine itself does not enter cells because of Pgp efflux, its octaarginine conjugate does so readily. This approach shows generality for overcoming the multidrug resistance elicited by small-molecule cancer chemotherapeutics and could improve the prognosis for many patients with cancer and fundamentally alter search strategies for novel therapeutic agents that are effective against resistant disease.


Assuntos
Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Oligopeptídeos/farmacologia , Paclitaxel/farmacocinética , Transporte Biológico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Oligopeptídeos/química , Oligopeptídeos/farmacocinética , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA