Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38139334

RESUMEN

As a substitution for hormone replacement therapy, many breast cancer patients use black cohosh (BC) extracts in combination with doxorubicin (DOX)-based chemotherapy. In this study, we evaluated the viability and survival of BC- and DOX-treated MCF-7 cells. A preclinical model of MCF-7 xenografts was used to determine the influence of BC and DOX administration on tumor growth and metabolism. The number of apoptotic cells after incubation with both DOX and BC was significantly increased (~100%) compared to the control. Treatment with DOX altered the potential of MCF-7 cells to form colonies; however, coincubation with BC did not affect this process. In vivo, PET-CT imaging showed that combined treatment of DOX and BC induced a significant reduction in both metabolic activity (29%) and angiogenesis (32%). Both DOX and BC treatments inhibited tumor growth by 20% and 12%, respectively, and combined by 57%, vs. control. We successfully demonstrated that BC increases cytotoxic effects of DOX, resulting in a significant reduction in tumor size. Further studies regarding drug transport and tumor growth biomarkers are necessary to establish the underlying mechanism and potential clinical use of BC in breast cancer patients.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Cimicifuga , Humanos , Femenino , Tomografía Computarizada por Tomografía de Emisión de Positrones , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Antineoplásicos/uso terapéutico , Células MCF-7 , Línea Celular Tumoral
2.
Cell Mol Biol Lett ; 26(1): 10, 2021 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-33726678

RESUMEN

BACKGROUND: Molecular imaging with molecularly targeted probes is a powerful tool for studying the spatio-temporal interactions between complex biological processes. The pivotal role of the receptor for advanced glycation end products (RAGE), and its involvement in numerous pathological processes, aroused the demand for RAGE-targeted imaging in various diseases. In the present study, we evaluated the use of a diagnostic imaging agent for RAGE quantification in an animal model of peripheral artery disease, a multimodal dual-labeled probe targeted at RAGE (MMIA-CML). METHODS: PAMAM dendrimer was conjugated with Nε-carboxymethyl-lysine (CML) modified albumin to synthesize the RAGE-targeted probe. A control untargeted agent carried native non-modified human albumin (HSA). Bifunctional p-SCN-Bn-NOTA was used to conjugate the 64Cu radioisotope. Surgical right femoral artery ligation was performed on C57BL/6 male mice. One week after femoral artery ligation, mice were injected with MMIA-CML or MMIA-HSA labeled with 64Cu radioisotope and 60 min later in vivo microPET-CT imaging was performed. Immediately after PET imaging studies, the murine hindlimb muscle tissues were excised and prepared for gene and protein expression analysis. RAGE gene and protein expression was assessed using real-time qPCR and Western blot technique respectively. To visualize RAGE expression in excised tissues, microscopic fluorescence imaging was performed using RAGE-specific antibodies and RAGE-targeted and -control MMIA. RESULTS: Animals subjected to PET imaging exhibited greater MMIA-CML uptake in ischemic hindlimbs than non-ischemic hindlimbs. We observed a high correlation between fluorescent signal detection and radioactivity measurement. Significant RAGE gene and protein overexpression were observed in ischemic hindlimbs compared to non-ischemic hindlimbs at one week after surgical ligation. Fluorescence microscopic staining revealed significantly increased uptake of RAGE-targeted nanoparticles in both ischemic and non-ischemic muscle tissues compared to the control probe but at a higher level in ischemic hindlimbs. Ischemic tissue exhibited explicit RAGE dyeing following anti-RAGE antibody and high colocalization with the MMIA-CML targeted at RAGE. CONCLUSIONS: The present results indicate increased expression of RAGE in the ischemic hindlimb and enable the use of multimodal nanoparticles in both in vitro and in vivo experimental models, creating the possibility for imaging structural and functional changes with a RAGE-targeted tracer.


Asunto(s)
Nanopartículas/química , Enfermedad Arterial Periférica/metabolismo , Enfermedad Arterial Periférica/patología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Animales , Modelos Animales de Enfermedad , Fluorescencia , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones
3.
Eur J Nucl Med Mol Imaging ; 47(11): 2562-2576, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32166512

RESUMEN

PURPOSE: Current screening and monitoring of prostate cancer (PCa) is insufficient, producing inaccurate diagnoses. Presence of the receptor for advanced glycation end-products (RAGE) is associated with signature characteristics of PCa development such as cell proliferation, anchorage-independent growth, angiogenesis, migration, invasion, and poor patient survival. Therefore, we developed a preclinical multimodal imaging strategy targeted at RAGE to diagnose and monitor PCa. METHODS: In this work, RAGE-targeted multimodal nanoparticles (64Cu-Cy5-G4-CML) were synthesized and rendered functional for nuclear and optical imaging using previously established methods. The probe's binding affinity and targeting specificity was assessed in androgen-dependent (LNCaP) and androgen-independent (DU145) prostate cancer cells using flow cytometry and confocal microscopy. In vivo PET-CT imaging was used to evaluate RAGE levels in DU145 and LNCaP xenograft models in mice. Then, tumors were excised post-imaging for histological staining and autoradiography to further assess RAGE levels and targeting efficiency of the tracer. Finally, RAGE levels from human PCa samples of varying Gleason Scores were evaluated using Western blot and immunohistochemical staining. RESULTS: PCa cell culture studies confirmed adequate RAGE-targeting with 64Cu-Cy5-G4-CML with KD between 360 and 540 nM as measured by flow cytometry. In vivo PET-CT images of PCa xenografts revealed favorable kinetics, rapid blood clearance, and a non-homogenous, enhanced uptake in tumors, which varied based on cell type and tumor size with mean uptake between 0.5 and 1.4%ID/g. RAGE quantification of human samples confirmed increased RAGE uptake corresponding to increased Gleason scoring. CONCLUSIONS: Our study has shown that RAGE-targeted cancer imaging is feasible and could significantly impact PCa management.


Asunto(s)
Radioisótopos de Cobre , Neoplasias de la Próstata , Animales , Humanos , Masculino , Ratones , Tomografía Computarizada por Tomografía de Emisión de Positrones , Neoplasias de la Próstata/diagnóstico por imagen , Receptor para Productos Finales de Glicación Avanzada
4.
Dev Cell ; 52(6): 748-763.e6, 2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-32109384

RESUMEN

Myotonic dystrophy type 1 (DM1) is a multisystemic genetic disorder caused by the CTG repeat expansion in the 3'-untranslated region of DMPK gene. Heart dysfunctions occur in ∼80% of DM1 patients and are the second leading cause of DM1-related deaths. Herein, we report that upregulation of a non-muscle splice isoform of RNA-binding protein RBFOX2 in DM1 heart tissue-due to altered splicing factor and microRNA activities-induces cardiac conduction defects in DM1 individuals. Mice engineered to express the non-muscle RBFOX240 isoform in heart via tetracycline-inducible transgenesis, or CRISPR/Cas9-mediated genome editing, reproduced DM1-related cardiac conduction delay and spontaneous episodes of arrhythmia. Further, by integrating RNA binding with cardiac transcriptome datasets from DM1 patients and mice expressing the non-muscle RBFOX2 isoform, we identified RBFOX240-driven splicing defects in voltage-gated sodium and potassium channels, which alter their electrophysiological properties. Thus, our results uncover a trans-dominant role for an aberrantly expressed RBFOX240 isoform in DM1 cardiac pathogenesis.


Asunto(s)
Potenciales de Acción , Frecuencia Cardíaca , Distrofia Miotónica/genética , Factores de Empalme de ARN/genética , Empalme del ARN , Proteínas Represoras/genética , Adulto , Animales , Células Cultivadas , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Distrofia Miotónica/metabolismo , Distrofia Miotónica/fisiopatología , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Empalme de ARN/metabolismo , Proteínas Represoras/metabolismo , Canales de Sodio Activados por Voltaje/genética , Canales de Sodio Activados por Voltaje/metabolismo
5.
Theranostics ; 10(4): 1733-1745, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32042333

RESUMEN

Background: Peripheral arterial disease (PAD) is a major worldwide health concern. Since the late 1990s therapeutic angiogenesis has been investigated as an alternative to traditional PAD treatments. Although positive preclinical results abound in the literature, the outcomes of human clinical trials have been discouraging. Among the challenges the field has faced has been a lack of standardization of the timings and measures used to validate new treatment approaches. Methods: In order to study the spatiotemporal dynamics of both perfusion and neovascularization in mice subjected to surgically-induced hindlimb ischemia (n= 30), we employed three label-free imaging modalities (a novel high-sensitivity ultrasonic Power Doppler methodology, laser speckle contrast, and photoacoustic imaging), as well as a tandem of radio-labeled molecular probes, 99mTc-NC100692 and 99mTc-BRU-5921 respectively, designed to detect two key modulators of angiogenic activity, αVß3 and HIF-1α , via scintigraphic imaging. Results: The multimodal imaging strategy reveals a set of "landmarks"-key physiological and molecular events in the healing process-that can serve as a standardized framework for describing the impact of emerging PAD treatments. These landmarks span the entire process of neovascularization, beginning with the rapid decreases in perfusion and oxygenation associated with ligation surgery, extending through pro-angiogenic changes in gene expression driven by the master regulator HIF-1α , and ultimately leading to complete functional revascularization of the affected tissues. Conclusions: This study represents an important step in the development of multimodal non-invasive imaging strategies for vascular research; the combined results offer more insight than can be gleaned through any of the individual imaging methods alone. Researchers adopting similar imaging strategies and will be better able to describe changes in the onset, duration, and strength of each of the landmarks of vascular recovery, yielding greater biological insight, and enabling more comprehensive cross-study comparisons. Perhaps most important, this study paves the road for more efficient translation of PAD research; emerging experimental treatments can be more effectively assessed and refined at the preclinical stage, ultimately leading to better next-generation therapies.


Asunto(s)
Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Imagen Multimodal/métodos , Enfermedad Arterial Periférica/terapia , Inductores de la Angiogénesis/metabolismo , Animales , Modelos Animales de Enfermedad , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Imidazoles , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/genética , Compuestos de Organotecnecio , Péptidos Cíclicos , Imagen de Perfusión/métodos , Enfermedad Arterial Periférica/diagnóstico por imagen , Técnicas Fotoacústicas/métodos , Cintigrafía/métodos , Recuperación de la Función , Ultrasonografía Doppler/métodos
6.
Theranostics ; 8(18): 5012-5024, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30429883

RESUMEN

The receptor for advanced glycation end-products (RAGE) is central to multiple disease states, including diabetes-related conditions such as peripheral arterial disease (PAD). Despite RAGE's importance in these pathologies, there remains a need for a molecular imaging agent that can accurately assess RAGE levels in vivo. Therefore, we have developed a multimodal nanoparticle-based imaging agent targeted at RAGE with the well-characterized RAGE ligand, carboxymethyllysine (CML)-modified human serum albumin (HSA). Methods: A multimodal tracer (64Cu-Rho-G4-CML) was developed using a generation-4 (G4) polyamidoamine (PAMAM) dendrimer, conjugated with both rhodamine and copper-64 (64Cu) chelator (NOTA) for optical and PET imaging, respectively. First, 64Cu-Rho-G4-CML and its non-targeted analogue (64Cu-Rho-G4-HSA) were evaluated chemically using techniques such as dynamic light scattering (DLS), electron microscopy and nuclear magnetic resonance (NMR). The tracers' binding capabilities were examined at the cellular level and optimized using live and fixed HUVEC cells grown in 5.5-30 mM glucose, followed by in vivo PET-CT imaging, where the probes' kinetics, biodistribution, and RAGE targeting properties were examined in a murine model of hindlimb ischemia. Finally, histological assessment of RAGE levels in both ischemic and non-ischemic tissues was performed. Conclusions: Our RAGE-targeted probe demonstrated an average size of 450 nm, a Kd of 340-390 nM, rapid blood clearance, and a 3.4 times greater PET uptake in ischemic RAGE-expressing hindlimbs than their non-ischemic counterpart. We successfully demonstrated increased RAGE expression in a murine model of hindlimb ischemia and the feasibility for non-invasive examination of cellular, tissue, and whole-body RAGE levels with a molecularly targeted tracer.


Asunto(s)
Antígenos de Neoplasias/análisis , Productos Finales de Glicación Avanzada/metabolismo , Lisina/análogos & derivados , Proteínas Quinasas Activadas por Mitógenos/análisis , Imagen Molecular/métodos , Imagen Multimodal/métodos , Nanopartículas/metabolismo , Albúmina Sérica Humana/metabolismo , Animales , Radioisótopos de Cobre/administración & dosificación , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lisina/metabolismo , Ratones
7.
Artículo en Inglés | MEDLINE | ID: mdl-30183625

RESUMEN

Combinations of novel pulse-echo acquisitions and clutter filtering techniques can improve the sensitivity and the specificity of power Doppler (PD) images, thus reducing the need for exogenous contrast enhancement. We acquire echoes following bursts of Doppler pulse transmissions sparsely applied in regular patterns over long durations. The goal is to increase the sensitivity of the acquisition to slow disorganized patterns of motion from the peripheral blood perfusion. To counter a concomitant increase in clutter signal power, we arrange the temporal echo acquisitions into two data-array axes, combine them with a spatial axis for the tissue region of interest, and apply 3-D singular-value decomposition (SVD) clutter filtering. Successful separation of blood echoes from other echo signal sources requires that we partition the 3-D SVD core tensor. Unfortunately, the clutter and blood subspaces do not completely uncouple in all situations, so we developed a statistical classifier that identifies the core tensor subspace dominated by tissue clutter power. This paper describes an approach to subspace partitioning as required for optimizing PD imaging of peripheral perfusion. The technique is validated using echo simulation, flow-phantom data, and in vivo data from a murine melanoma model. We find that for narrow eigen-bandwidth clutter signals, we can routinely map phantom flows and tumor perfusion signals at speeds less than 3 mL/min. The proposed method is well suited to peripheral perfusion imaging applications.


Asunto(s)
Imagen de Perfusión/métodos , Procesamiento de Señales Asistido por Computador , Ultrasonografía/métodos , Algoritmos , Animales , Velocidad del Flujo Sanguíneo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales , Fantasmas de Imagen
8.
ACS Cent Sci ; 4(8): 1045-1055, 2018 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-30159402

RESUMEN

Cancer stem cells (CSCs) are progenitor cells that contribute to treatment-resistant phenotypes during relapse. CSCs exist in specific tissue microenvironments that cell cultures and more complex models cannot mimic. Therefore, the development of new approaches that can detect CSCs and report on specific properties (e.g., stem cell plasticity) in their native environment have profound implications for studying CSC biology. Herein, we present AlDeSense, a turn-on fluorescent probe for aldehyde dehydrogenase 1A1 (ALDH1A1) and Ctrl-AlDeSense, a matching nonresponsive reagent. Although ALDH1A1 contributes to the detoxification of reactive aldehydes, it is also associated with stemness and is highly elevated in CSCs. AlDeSense exhibits a 20-fold fluorescent enhancement when treated with ALDH1A1. Moreover, we established that AlDeSense is selective against a panel of common ALDH isoforms and exhibits exquisite chemostability against a collection of biologically relevant species. Through the application of surface marker antibody staining, tumorsphere assays, and assessment of tumorigenicity, we demonstrate that cells exhibiting high AlDeSense signal intensity have properties of CSCs. Using these probes in tandem, we have identified CSCs at the cellular level via flow cytometry and confocal imaging, as well as monitored their states in animal models.

9.
Sci Rep ; 8(1): 5401, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29599497

RESUMEN

The α V ß3 integrin plays an important role in many physiological functions and pathological disorders. α V ß3 is minimally expressed in normal quiescent endothelial cells, but significantly upregulated during neovascularization. In this study, we evaluated a 64Cu-labeled dimeric cRGD tracer targeted at α V ß3 integrin and report its applicability to assess peripheral angiogenesis in diabetes mellitus (DM). We established a murine model of type-1 DM characterized by elevated glucose, glycated serum protein (GSP), and glycated hemoglobin A1c (HbA1c). We demonstrated that our imaging probe is specific to α V ß3 integrin under both normo- and hyperglycemic conditions. We found that the analysis of in vivo PET-CT images correlated well with gamma well counting (GWC). Both GWC and PET-CT imaging demonstrated increased uptake of 64Cu-NOTA-PEG4-cRGD2 in the ischemic hindlimb in contrast to non-ischemic control. GWC of the distal ischemic tissue from DM mice showed significantly lower probe accumulation than in non-DM mice. The immunofluorescence staining of the ischemic tissues showed a 3-fold reduction in CD31 and 4-fold reduction in the α V ß3 expression in DM vs. non-DM animals. In conclusion, we successfully demonstrated that diabetes-associated reductions in peripheral angiogenesis can be non-invasively detected with PET-CT imaging using targeted dimeric-cRGD probe.


Asunto(s)
Diabetes Mellitus Experimental/patología , Neovascularización Fisiológica , Péptidos Cíclicos/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Animales , Biomarcadores/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/diagnóstico por imagen , Dimerización , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfaVbeta3/química , Integrina alfaVbeta3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Péptidos Cíclicos/química , Unión Proteica , Radiofármacos/química , Radiofármacos/metabolismo , Distribución Tisular
10.
Nat Commun ; 8(1): 1794, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-29176550

RESUMEN

Hypoxia occurs when limited oxygen supply impairs physiological functions and is a pathological hallmark of many diseases including cancer and ischemia. Thus, detection of hypoxia can guide treatment planning and serve as a predictor of patient prognosis. Unfortunately, current methods suffer from invasiveness, poor resolution and low specificity. To address these limitations, we present Hypoxia Probe 1 (HyP-1), a hypoxia-responsive agent for photoacoustic imaging. This emerging modality converts safe, non-ionizing light to ultrasound waves, enabling acquisition of high-resolution 3D images in deep tissue. HyP-1 features an N-oxide trigger that is reduced in the absence of oxygen by heme proteins such as CYP450 enzymes. Reduction of HyP-1 produces a spectrally distinct product, facilitating identification via photoacoustic imaging. HyP-1 exhibits selectivity for hypoxic activation in vitro, in living cells, and in multiple disease models in vivo. HyP-1 is also compatible with NIR fluorescence imaging, establishing its versatility as a multimodal imaging agent.


Asunto(s)
Diagnóstico por Imagen/métodos , Hipoxia/diagnóstico por imagen , Óxidos/química , Técnicas Fotoacústicas/métodos , Animales , Línea Celular Tumoral , Femenino , Humanos , Imagenología Tridimensional/métodos , Isquemia/diagnóstico por imagen , Isquemia/patología , Ratones , Ratones Endogámicos BALB C , Microsomas Hepáticos , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Oxidación-Reducción , Oxígeno/metabolismo , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Theranostics ; 7(16): 3876-3888, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29109784

RESUMEN

Peripheral arterial disease (PAD) is a debilitating complication of diabetes mellitus (DM) that leads to thousands of injuries, amputations, and deaths each year. The use of mesenchymal stem cells (MSCs) as a regenerative therapy holds the promise of regrowing injured vasculature, helping DM patients live healthier and longer lives. We report the use of muscle-derived MSCs to treat surgically-induced hindlimb ischemia in a mouse model of type 1 diabetes (DM-1). We serially evaluate several facets of the recovery process, including αVß3 -integrin expression (a marker of angiogenesis), blood perfusion, and muscle function. We also perform microarray transcriptomics experiments to characterize the gene expression states of the MSC-treated is- chemic tissues, and compare the results with those of non-ischemic tissues, as well as ischemic tissues from a saline-treated control group. The results show a multifaceted impact of mMSCs on hindlimb ischemia. We determined that the angiogenic activity one week after mMSC treatment was enhanced by approximately 80% relative to the saline group, which resulted in relative increases in blood perfusion and muscle strength of approximately 42% and 1.7-fold, respectively. At the transcriptomics level, we found that several classes of genes were affected by mMSC treatment. The mMSCs appeared to enhance both pro-angiogenic and metabolic genes, while suppressing anti-angiogenic genes and certain genes involved in the inflammatory response. All told, mMSC treatment appears to exert far-reaching effects on the microenvironment of ischemic tissue, enabling faster and more complete recovery from vascular occlusion.


Asunto(s)
Angiopatías Diabéticas/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Animales , Angiopatías Diabéticas/complicaciones , Angiopatías Diabéticas/diagnóstico por imagen , Angiopatías Diabéticas/fisiopatología , Regulación de la Expresión Génica , Procesamiento de Imagen Asistido por Computador , Integrina alfaVbeta3/metabolismo , Isquemia/patología , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Músculos/fisiopatología , Neovascularización Fisiológica , Perfusión , Enfermedad Arterial Periférica/complicaciones , Enfermedad Arterial Periférica/patología , Enfermedad Arterial Periférica/terapia , Tomografía Computarizada por Tomografía de Emisión de Positrones , Cambios Post Mortem , Proteoma/metabolismo , Distribución Tisular , Transcriptoma/genética
12.
Sci Adv ; 3(10): e1701350, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29075670

RESUMEN

Tumor angiogenesis provides critical nutrients for cancer progression and may also facilitate pathways for dissemination during the process of metastasis. It is well established that cells that metastasize display characteristics of stem cells; however, the prevailing paradigm points to these stem-like cells residing in the hypoxic niche within the tumor interior. Controlling the geometry at the interface of a population of melanoma cells reveals a role for perimeter topology in promoting a stem-like state with enhanced tumorigenicity. We show that this putative melanoma-initiating cell (MIC) demonstrates significant enhancement in the secretion of proangiogenic molecules. This finding suggests the possibility of an "invasive niche" at the perimeter of a growing tumor that promotes a MIC state with angiogenic activity. Using several in vitro and in vivo models of tumor angiogenesis, we see concurrent stem-like characteristics with initiation of neovascularization. In the absence of hypoxia, precise topological cues induce signaling through integrin α5ß1 and downstream extracellular signal-regulated kinase (ERK) signaling to regulate the MIC secretome through the signal transducer and activator of transcription (STAT) and hypoxia-inducible factor 1α (HIF1α) pathways. Inhibiting integrin α5ß1 and ERK signaling attenuates both the MIC phenotype and proangiogenic signaling. These results suggest that topological cues in the periphery of malignant melanoma promote the MIC state-using mechanotransduction in lieu of low oxygen-to facilitate the formation of new vasculature for progression and invasion.


Asunto(s)
Melanoma/metabolismo , Melanoma/patología , Células Madre Neoplásicas/inmunología , Neovascularización Patológica/metabolismo , Fenotipo , Animales , Biomarcadores , Adhesión Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Citocinas/metabolismo , Citoesqueleto/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunofenotipificación , Integrinas/metabolismo , Mecanotransducción Celular , Melanoma/diagnóstico por imagen , Melanoma Experimental , Ratones , Imagen Molecular , Células Madre Neoplásicas/patología
13.
Sci Rep ; 7(1): 3185, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28600529

RESUMEN

Cyclic peptides containing the Arg-Gly-Asp (RGD) sequence have been shown to specifically bind the angiogenesis biomarker α V ß 3 integrin. We report the synthesis, chemical characterization, and biological evaluation of two novel dimeric cyclic RGD-based molecular probes for the targeted imaging of α V ß 3 activity (a radiolabeled version, 64Cu-NOTA-PEG4-cRGD2, for PET imaging, and a fluorescent version, FITC-PEG4-cRGD2, for in vitro work). We investigated the performance of this probe at the receptor, cell, organ, and whole-body levels, including its use to detect diabetes associated impairment of ischemia-induced myocardial angiogenesis. Both versions of the probe were found to be stable, demonstrated fast receptor association constants, and showed high specificity for α V ß 3 in HUVECs (K d ~ 35 nM). Dynamic PET-CT imaging indicated rapid blood clearance via kidney filtration, and accumulation within α V ß 3-positive infarcted myocardium. 64Cu-NOTA-PEG4-cRGD2 demonstrated a favorable biodistribution, slow washout, and excellent performance with respect to the quality of the PET-CT images obtained. Importantly, the ratio of probe uptake in infarcted heart tissue compared to normal tissue was significantly higher in non-diabetic rats than in diabetic ones. Overall, our probes are promising agents for non-invasive quantitative imaging of α V ß 3 expression, both in vitro and in vivo.


Asunto(s)
Integrina alfaVbeta3/genética , Neovascularización Patológica/tratamiento farmacológico , Péptidos Cíclicos/farmacología , Animales , Línea Celular Tumoral , Radioisótopos de Cobre/farmacología , Dimerización , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos con 1 Anillo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Riñón/efectos de los fármacos , Riñón/metabolismo , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Oligopéptidos/química , Oligopéptidos/farmacología , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/química , Tomografía de Emisión de Positrones , Ratas , Distribución Tisular/efectos de los fármacos
14.
Artículo en Inglés | MEDLINE | ID: mdl-28650810

RESUMEN

A method is explored for increasing the sensitivity of power-Doppler imaging without contrast enhancement. We acquire 1-10 s of echo signals and arrange it into a 3-D spatiotemporal data array. An eigenfilter developed to preserve all three dimensions of the array yields power estimates for blood flow and perfusion that are well separated from tissue clutter. This method is applied at high frequency (24-MHz pulses) to a murine model of an ischemic hindlimb. We demonstrate enhancements to tissue perfusion maps in normal and ischemic tissues. The method can be applied to data from any ultrasonic instrument that provides beamformed RF echo data.


Asunto(s)
Algoritmos , Velocidad del Flujo Sanguíneo/fisiología , Procesamiento de Imagen Asistido por Computador/métodos , Imagen de Perfusión/métodos , Ultrasonografía/métodos , Animales , Miembro Posterior/irrigación sanguínea , Miembro Posterior/diagnóstico por imagen , Isquemia/diagnóstico por imagen , Ratones
15.
BMC Syst Biol ; 9: 15, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25890263

RESUMEN

BACKGROUND: The exchange of metabolites and the reprogramming of metabolism in response to shifting microenvironmental conditions can drive subpopulations of cells within colonies toward divergent behaviors. Understanding the interactions of these subpopulations-their potential for competition as well as cooperation-requires both a metabolic model capable of accounting for a wide range of environmental conditions, and a detailed dynamic description of the cells' shared extracellular space. RESULTS: Here we show that a cell's position within an in silico Escherichia coli colony grown on glucose minimal agar can drastically affect its metabolism: "pioneer" cells at the outer edge engage in rapid growth that expands the colony, while dormant cells in the interior separate two spatially distinct subpopulations linked by a cooperative form of acetate crossfeeding that has so far gone unnoticed. Our hybrid simulation technique integrates 3D reaction-diffusion modeling with genome-scale flux balance analysis (FBA) to describe the position-dependent metabolism and growth of cells within a colony. Our results are supported by imaging experiments involving strains of fluorescently-labeled E. coli. The spatial patterns of fluorescence within these experimental colonies identify cells with upregulated genes associated with acetate crossfeeding and are in excellent agreement with the predictions. Furthermore, the height-to-width ratios of both the experimental and simulated colonies are in good agreement over a growth period of 48 hours. CONCLUSIONS: Our modeling paradigm can accurately reproduce a number of known features of E. coli colony growth, as well as predict a novel one that had until now gone unrecognized. The acetate crossfeeding we see has a direct analogue in a form of lactate crossfeeding observed in certain forms of cancer, and we anticipate future application of our methodology to models of tissues and tumors.


Asunto(s)
Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Modelos Biológicos , Proliferación Celular , Simulación por Computador , Difusión , Escherichia coli/citología , Análisis de Flujos Metabólicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...