Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Stem Cells ; 33(4): 1241-53, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25640064

RESUMEN

Animal studies have shown that mesenchymal stromal cell (MSC) infusions improve acute kidney injury (AKI) outcomes when administered early after ischemic/reperfusion injury or within 24 hours after cisplatin administration. These findings have spurred several human clinical trials to prevent AKI. However, no specific therapy effectively treats clinically obvious AKI or rescues renal function once advanced injury is established. We investigated if noninvasive image-guided pulsed focused ultrasound (pFUS) could alter the kidney microenvironment to enhance homing of subsequently infused MSC. To examine the efficacy of pFUS-enhanced cell homing in disease, we targeted pFUS to kidneys to enhance MSC homing after cisplatin-induced AKI. We found that pFUS enhanced MSC homing at 1 day post-cisplatin, prior to renal functional deficits, and that enhanced homing improved outcomes of renal function, tubular cell death, and regeneration at 5 days post-cisplatin compared to MSC alone. We then investigated whether pFUS+MSC therapy could rescue established AKI. MSC alone at 3 days post-cisplatin, after renal functional deficits were obvious, significantly improved 7-day survival of animals. Survival was further improved by pFUS and MSC. pFUS prior to MSC injections increased IL-10 production by MSC that homed to kidneys and generated an anti-inflammatory immune cell profile in treated kidneys. This study shows pFUS is a neoadjuvant approach to improve MSC homing to diseased organs. pFUS with MSC better prevents AKI than MSC alone and allows rescue therapy in established AKI, which currently has no meaningful therapeutic options.


Asunto(s)
Lesión Renal Aguda/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Ondas Ultrasónicas , Lesión Renal Aguda/patología , Animales , Femenino , Humanos , Células Madre Mesenquimatosas/efectos de la radiación , Ratones , Ratones Endogámicos C3H , Resultado del Tratamiento
2.
Stem Cells ; 31(11): 2551-60, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23922277

RESUMEN

Stem cells are promising therapeutics for cardiovascular diseases, and i.v. injection is the most desirable route of administration clinically. Subsequent homing of exogenous stem cells to pathological loci is frequently required for therapeutic efficacy and is mediated by chemoattractants (cell adhesion molecules, cytokines, and growth factors). Homing processes are inefficient and depend on short-lived pathological inflammation that limits the window of opportunity for cell injections. Noninvasive pulsed focused ultrasound (pFUS), which emphasizes mechanical ultrasound-tissue interactions, can be precisely targeted in the body and is a promising approach to target and maximize stem cell delivery by stimulating chemoattractant expression in pFUS-treated tissue prior to cell infusions. We demonstrate that pFUS is nondestructive to murine skeletal muscle tissue (no necrosis, hemorrhage, or muscle stem cell activation) and initiates a largely M2-type macrophage response. We also demonstrate that local upregulation of chemoattractants in pFUS-treated skeletal muscle leads to enhance homing, permeability, and retention of human mesenchymal stem cells (MSC) and human endothelial precursor cells (EPC). Furthermore, the magnitude of MSC or EPC homing was increased when pFUS treatments and cell infusions were repeated daily. This study demonstrates that pFUS defines transient "molecular zip codes" of elevated chemoattractants in targeted muscle tissue, which effectively provides spatiotemporal control and tunability of the homing process for multiple stem cell types. pFUS is a clinically translatable modality that may ultimately improve homing efficiency and flexibility of cell therapies for cardiovascular diseases.


Asunto(s)
Citocinas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Músculo Esquelético/metabolismo , Células Madre/metabolismo , Ultrasonido/métodos , Animales , Técnicas de Cultivo de Célula , Femenino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/diagnóstico por imagen , Ratones , Ratones Endogámicos C3H , Modelos Animales , Músculo Esquelético/citología , Músculo Esquelético/diagnóstico por imagen , Análisis Espacio-Temporal , Células Madre/citología , Células Madre/diagnóstico por imagen , Ultrasonografía
3.
J Acoust Soc Am ; 133(3): 1827-34, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23464051

RESUMEN

Gene therapy by intratumoral injection is a promising approach for treating solid tumors. However, this approach has limited success due to insufficient distribution of gene vectors used for gene delivery. Previous studies have shown that pulsed-focused ultrasound (pFUS) can enhance both systemic and local delivery of therapeutic agents in solid tumors and other disease models. Here, murine squamous cell carcinoma flank tumors were treated with single intratumoral injection of naked tumor necrosis factor-alpha (TNF-α) plasmid, either with or without a preceding pFUS exposure. The exposures were given at 1 MHz, at a spatial average, temporal peak intensity of 2660 W cm(-2), using 50 ms pulses, given at a pulse repetition frequency of 1 Hz. One hundred pulses were given at individual raster points, spaced evenly over the projected surface of the tumor at a distance of 2 mm. Exposures alone had no effect on tumor growth. Significant growth inhibition was observed with injection of TNF-α plasmid, and tumor growth was further inhibited with pFUS. Improved results with pFUS correlated with larger necrotic regions in histological sections and improved distribution and penetration of fluorescent surrogate nanoparticles. Electron microscopy demonstrated enlarged gaps between cells in exposed tissue, and remote acoustic palpation showed decreases in tissue stiffness after pFUS. Combined, these results suggest pFUS effects may be reducing barriers for tissue transport and additionally lowering interstitial fluid pressure to further improve delivery and distribution of injected plasmid for greater therapeutic effects. This suggests that pFUS could potentially be beneficial for improving local gene therapy treatment of human malignancies.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Terapia Genética , Neoplasias de los Tejidos Blandos/terapia , Factor de Necrosis Tumoral alfa/biosíntesis , Ultrasonido , Acústica , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/ultraestructura , Femenino , Humanos , Ratones , Ratones Endogámicos C3H , Microscopía Electrónica de Transmisión , Necrosis , Palpación , Permeabilidad , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/inmunología , Neoplasias de los Tejidos Blandos/ultraestructura , Tejido Subcutáneo/inmunología , Tejido Subcutáneo/ultraestructura , Factores de Tiempo , Carga Tumoral , Factor de Necrosis Tumoral alfa/genética
4.
Stem Cells ; 30(6): 1216-27, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22593018

RESUMEN

Bone marrow stromal cells (BMSCs) have shown significant promise in the treatment of disease, but their therapeutic efficacy is often limited by inefficient homing of systemically administered cells, which results in low number of cells accumulating at sites of pathology. BMSC home to areas of inflammation where local expression of integrins and chemokine gradients is present. We demonstrated that nondestructive pulsed focused ultrasound (pFUS) exposures that emphasize the mechanical effects of ultrasound-tissue interactions induced local and transient elevations of chemoattractants (i.e., cytokines, integrins, and growth factors) in the murine kidney. pFUS-induced upregulation of cytokines occurred through approximately 1 day post-treatment and returned to contralateral kidney levels by day 3. This window of significant increases in cytokine expression was accompanied by local increases of other trophic factors and integrins that have been shown to promote BMSC homing. When BMSCs were intravenously administered following pFUS treatment to a single kidney, enhanced homing, permeability, and retention of BMSC was observed in the treated kidney versus the contralateral kidney. Histological analysis revealed up to eight times more BMSC in the peritubular regions of the treated kidneys on days 1 and 3 post-treatment. Furthermore, cytokine levels in pFUS-treated kidneys following BMSC administration were found to be similar to controls, suggesting modulation of cytokine levels by BMSC. pFUS could potentially improve cell-based therapies as a noninvasive modality to target homing by establishing local chemoattractant gradients and increasing expression of integrins to enhance tropism of cells toward treated tissues.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/diagnóstico por imagen , Trasplante de Médula Ósea/métodos , Riñón/citología , Riñón/diagnóstico por imagen , Células del Estroma/trasplante , Ultrasonido/métodos , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea/diagnóstico por imagen , Técnicas de Cultivo de Célula , Citocinas/metabolismo , Femenino , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/diagnóstico por imagen , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células del Estroma/citología , Ultrasonografía
5.
PLoS One ; 6(9): e24730, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931834

RESUMEN

Continuous focused ultrasound (cFUS) has been widely used for thermal ablation of tissues, relying on continuous exposures to generate temperatures necessary to induce coagulative necrosis. Pulsed FUS (pFUS) employs non-continuous exposures that lower the rate of energy deposition and allow cooling to occur between pulses, thereby minimizing thermal effects and emphasizing effects created by non-thermal mechanisms of FUS (i.e., acoustic radiation forces and acoustic cavitation). pFUS has shown promise for a variety of applications including drug and nanoparticle delivery; however, little is understood about the effects these exposures have on tissue, especially with regard to cellular pro-homing factors (growth factors, cytokines, and cell adhesion molecules). We examined changes in murine hamstring muscle following pFUS or cFUS and demonstrate that pFUS, unlike cFUS, has little effect on the histological integrity of muscle and does not induce cell death. Infiltration of macrophages was observed 3 and 8 days following pFUS or cFUS exposures. pFUS increased expression of several cytokines (e.g., IL-1α, IL-1ß, TNFα, INFγ, MIP-1α, MCP-1, and GMCSF) creating a local cytokine gradient on days 0 and 1 post-pFUS that returns to baseline levels by day 3 post-pFUS. pFUS exposures induced upregulation of other signaling molecules (e.g., VEGF, FGF, PlGF, HGF, and SDF-1α) and cell adhesion molecules (e.g., ICAM-1 and VCAM-1) on muscle vasculature. The observed molecular changes in muscle following pFUS may be utilized to target cellular therapies by increasing homing to areas of pathology.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación , Animales , Apoptosis/fisiología , Moléculas de Adhesión Celular/metabolismo , Quimiocina CCL3/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1/metabolismo , Interleucina-1beta/metabolismo , Macrófagos , Imagen por Resonancia Magnética , Ratones , Músculo Esquelético/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
6.
Magn Reson Imaging ; 28(3): 320-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20061111

RESUMEN

PURPOSE: The objective of this study was to evaluate diffusion anisotropy of the breast parenchyma and assess the range and repeatability of diffusion tensor imaging (DTI) parameters in normal breast tissue. MATERIALS AND METHODS: The study was approved by our institutional review board and included 12 healthy females (median age, 36 years). Diffusion tensor imaging was performed at 1.5 T using a diffusion-weighted echo planar imaging sequence. Diffusion tensor imaging parameters including tensor eigenvalues (lambda(1), lambda(2), lambda(3)), fractional anisotropy (FA) and apparent diffusion coefficient (ADC) were measured for anterior, central and posterior breast regions. RESULTS: Mean normal breast DTI measures were lambda(1)=2.51 x 10(-3) mm(2)/s, lambda(2)=1.89 x 10(-3) mm(2)/s, lambda(3)=1.39 x 10(-3) mm(2)/s, ADC=1.95+/-0.24 x 10(-3) mm(2)/s and FA=0.29+/-0.05 for b=600 s/mm(2). Significant regional differences were observed for both FA and ADC (P<.05), with higher ADC in the central breast and higher FA in the posterior breast. Comparison of DTI values calculated using b=0, 600 s/mm(2) vs. b=0, 1000 s/mm(2), showed significant differences in ADC (P<.001), but not FA. Repeatability assessment produced within-subject coefficient of variations of 4.5% for ADC and 11.4% for FA measures. CONCLUSION: This study demonstrates anisotropy of water diffusion in normal breast tissue and establishes a normative range of breast FA values. Attention to the influence of breast region and b value on breast DTI measurements may be important for clinical interpretation and standardization of techniques.


Asunto(s)
Mama/anatomía & histología , Mama/fisiología , Imagen de Difusión por Resonancia Magnética/métodos , Adulto , Anisotropía , Femenino , Humanos , Valores de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
7.
J Magn Reson Imaging ; 31(2): 339-47, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20099346

RESUMEN

PURPOSE: To investigate whether diffusion tensor imaging (DTI) measures of anisotropy in breast tumors are different from normal breast tissue and can improve the discrimination between benign and malignant lesions. MATERIALS AND METHODS: The study included 81 women with 105 breast lesions (76 malignant, 29 benign). DTI was performed during breast MRI examinations, and fractional anisotropy (FA) and apparent diffusion coefficient (ADC) values were measured for breast lesions and normal tissue in each subject. FA and ADC were compared between cancers, benign lesions, and normal tissue by univariate and multivariate analyses. RESULTS: The FA of carcinomas (mean +/- SD: 0.24 +/- 0.07) was significantly lower than normal breast tissue in the same subjects (0.29 +/- 0.07; P < 0.0001). Multiple logistic regression showed that FA and ADC were each independent discriminators of malignancy (P < 0.0001), and that FA improved discrimination between cancer and normal tissue over ADC alone. However, there was no difference in FA between malignant and benign lesions (P = 0.98). CONCLUSION: Diffusion anisotropy is significantly lower in breast cancers than normal tissue, which may reflect alterations in tissue organization. Our preliminary results suggest that FA adds incremental value over ADC alone for discriminating malignant from normal tissue but does not help with distinguishing benign from malignant lesions.


Asunto(s)
Algoritmos , Neoplasias de la Mama/diagnóstico , Imagen de Difusión por Resonancia Magnética/métodos , Interpretación de Imagen Asistida por Computador/métodos , Imagenología Tridimensional/métodos , Adulto , Anciano , Anciano de 80 o más Años , Anisotropía , Femenino , Humanos , Aumento de la Imagen/métodos , Persona de Mediana Edad , Proyectos Piloto , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Adulto Joven
8.
Artículo en Inglés | MEDLINE | ID: mdl-19163704

RESUMEN

Real-time three-dimensional ultrasound imaging (4D US) was utilized to monitor the treatment site during high-intensity focused ultrasound (HIFU) treatment. To obtain real-time monitoring during HIFU sonication, a 4D US imaging system and HIFU were synchronized and interference on the US image adjusted so that the region of interest was visible during treatment. The system was tested using tissue mimicking phantom gels and chicken breast tissue. The 4D US showed hyperechoic spots at the focal region of the HIFU transducer which then slowly faded after HIFU treatment. The hyperechoic regions were used as an indication of coagulative necrosis which occurs at temperatures higher than 60 degrees C. Different intensities of HIFU were applied to observe the difference in lesion formation and to determine the threshold intensity that produced hyperechoic regions due to the thermal and mechanical effects of focused ultrasound waves. The position, orientation, and shape of various lesions were examined in the three dimensional ultrasound images, and the volume of the lesions was measured. These volumes were compared to the volume measurements obtained from dissection of the tissue and phantom gels.


Asunto(s)
Imagenología Tridimensional/métodos , Ultrasonografía/métodos , Algoritmos , Animales , Calibración , Pollos , Diseño de Equipo , Glándulas Mamarias Animales/patología , Fantasmas de Imagen , Temperatura , Factores de Tiempo , Terapia por Ultrasonido , Ultrasonido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA