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











Base de datos
Intervalo de año de publicación
1.
Transl Oncol ; 14(1): 100937, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33217645

RESUMEN

For decades, sodium/iodide symporter NIS-mediated iodide uptake has played a crucial role in the radioactive ablation of thyroid cancer cells. NIS-based gene therapy has also become a promising tool for the treatment of tumors of extrathyroidal origin. But its applicability has been hampered by reduced expression of NIS, resulting in a moderated capacity to accumulate 131I and in inefficient ablation. Despite numerous preclinical enhancement strategies, the understanding of NIS expression within tumors remains limited. This study aims at a better understanding of the functional behavior of exogenous NIS expression in the context of malignant solid tumors that are characterized by rapid growth with an insufficient vasculature, leading to hypoxia and quiescence. Using subcutaneous HT29NIS and K7M2NIS tumors, we show that NIS-mediated uptake and NIS expression at the plasma membrane of cancer cells are impaired in the intratumoral regions. For a better understanding of the underlying molecular mechanisms induced by hypoxia and quiescence (separately and in combination), we performed experiments on HT29NIS cancer cells. Hypoxia and quiescence were both found to impair NIS-mediated uptake through mechanisms including NIS mis-localization. Modifications in the expression of proteins and metabolites involved in plasma membrane localization and in energy metabolism were found using untargeted proteomics and metabolomics approaches. In conclusion, our results provide evidence that hypoxia and quiescence impair NIS expression at the plasma membrane, and iodide uptake. Our study also shows that the tumor microenvironment is an important parameter for successful NIS-based cancer treatment.

2.
PLoS Pathog ; 16(10): e1008660, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33075093

RESUMEN

Mammary carcinoma, including triple-negative breast carcinomas (TNBC) are tumor-types for which human and canine pathologies are closely related at the molecular level. The efficacy of an oncolytic vaccinia virus (VV) was compared in low-passage primary carcinoma cells from TNBC versus non-TNBC. Non-TNBC cells were 28 fold more sensitive to VV than TNBC cells in which VV replication is impaired. Single-cell RNA-seq performed on two different TNBC cell samples, infected or not with VV, highlighted three distinct populations: naïve cells, bystander cells, defined as cells exposed to the virus but not infected and infected cells. The transcriptomes of these three populations showed striking variations in the modulation of pathways regulated by cytokines and growth factors. We hypothesized that the pool of genes expressed in the bystander populations was enriched in antiviral genes. Bioinformatic analysis suggested that the reduced activity of the virus was associated with a higher mesenchymal status of the cells. In addition, we demonstrated experimentally that high expression of one gene, DDIT4, is detrimental to VV production. Considering that DDIT4 is associated with a poor prognosis in various cancers including TNBC, our data highlight DDIT4 as a candidate resistance marker for oncolytic poxvirus therapy. This information could be used to design new generations of oncolytic poxviruses. Beyond the field of gene therapy, this study demonstrates that single-cell transcriptomics can be used to identify cellular factors influencing viral replication.


Asunto(s)
Neoplasias Mamarias Animales/metabolismo , Viroterapia Oncolítica/métodos , Factores de Transcripción/metabolismo , Transcriptoma , Virus Vaccinia/genética , Vaccinia/metabolismo , Replicación Viral , Animales , Biología Computacional , Perros , Femenino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/terapia , Neoplasias Mamarias Animales/virología , Análisis de la Célula Individual , Factores de Transcripción/genética , Vaccinia/genética , Vaccinia/virología
3.
Langmuir ; 36(35): 10460-10470, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32787032

RESUMEN

Diffusion of nanomedicines inside the extracellular matrix (ECM) has been identified as a key factor to achieve homogeneous distribution and therefore therapeutic efficacy. Here, we sought to determine the impact of nanoparticles' (NPs) surface properties on their ability to diffuse in the ECM. As model nano-objects, we used a library of gold nanoparticles grafted with a versatile polymethacrylate corona, which enabled the surface properties to be modified. To accurately recreate the features of the native ECM, diffusion studies were carried out in a tumor-derived gel (Matrigel). We developed two methods to evaluate the diffusion ability of NPs inside this model gel: an easy-to-implement one based on optical monitoring and another one using small-angle X-ray scattering (SAXS) measurements. Both enabled the determination of the diffusion coefficients of NPs and comparison of the influence of their various surface properties, while the SAXS technique also allowed to monitor the NPs' structure as they diffused inside the gel. Positive charges and hydrophobicity were found to particularly hinder diffusion, and the different results suggested on the whole the presence of NPs-matrix interactions, therefore underlying the importance of the ECM model. The accuracy of the tumor-derived gels used in this study was evidenced by in vivo experiments involving intratumoral injections of NPs on mice, which showed that diffusion patterns in the peripheral tumor tissues were quite similar to the ones obtained within the chosen ECM model.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Animales , Colágeno , Combinación de Medicamentos , Matriz Extracelular , Oro , Laminina , Ratones , Polímeros , Proteoglicanos , Dispersión del Ángulo Pequeño , Propiedades de Superficie , Difracción de Rayos X
4.
J Clin Med ; 9(2)2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31979418

RESUMEN

(1) Background: We recently showed that iodinated contrast media (ICM) reduced thyroid uptake of iodide independently of free iodide through a mechanism different from that of NaI and involving a dramatic and long-lasting decrease in Na/I symporter expression. The present study aimed at comparing the response of the thyroid to ICM and NaI using a quantitative proteomic approach. (2) Methods: Scintiscans were performed on ICM-treated patients. Micro Single-Photon Emission Computed Tomography (microSPECT/CT) imaging was used to assess thyroid uptakes in ICM- or NaI-treated mice and their response to recombinant human thyroid-stimulating hormone. Total thyroid iodide content and proteome was determined in control, NaI-, or ICM-treated animals. (3) Results: The inhibitory effect of ICM in patients was selectively observed on thyroids but not on salivary glands for up to two months after a systemic administration. An elevated level of iodide was observed in thyroids from NaI-treated mice but not in those from ICM animals. Exposure of the thyroid to NaI modulates 15 cellular pathways, most of which are also affected by ICM treatment (including the elF4 and P706SK cell signaling pathway and INSR identified as an upstream activator in both treatments). In addition, ICM modulates 16 distinct pathways and failed to affect thyroid iodide content. Finally, administration of ICM reduces thyroid-stimulating hormone (TSH) receptor expression which results in a loss of TSH-induced iodide uptake by the thyroid. (4) Conclusions: Common intracellular mechanisms are involved in the ICM- and NaI-induced reduction of iodide uptake. However, ICM fails to affect thyroid iodide content which suggests that the modulation of these common pathways is triggered by separate effectors. ICM also modulates numerous distinct pathways which may account for its long-lasting effect on thyroid uptake. These observations may have implications in the management of patients affected by differentiated thyroid carcinomas who have been exposed to ICM. They also provide the basis for the utilization of ICM-based compounds in radioprotection of the thyroid.

5.
Nanomedicine ; 23: 102084, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31454552

RESUMEN

Although chemically synthesized ferro/ferrimagnetic nanoparticles have attracted great attention in cancer theranostics, they lack radio-enhancement efficacy due to low targeting and internalization ability. Herein, we investigated the potential of RGD-tagged magnetosomes, bacterial biogenic magnetic nanoparticles naturally coated with a biological membrane and genetically engineered to express an RGD peptide, as tumor radioenhancers for conventional radiotherapy and proton therapy. Although native and RGD-magnetosomes similarly enhanced radiation-induced damage to plasmid DNA, RGD-magnetoprobes were able to boost the efficacy of radiotherapy to a much larger extent than native magnetosomes both on cancer cells and in tumors. Combined to magnetosomes@RGD, proton therapy exceeded the efficacy of X-rays at equivalent doses. Also, increased secondary emissions were measured after irradiation of magnetosomes with protons versus photons. Our results indicate the therapeutic advantage of using functionalized magnetoparticles to sensitize tumors to both X-rays and protons and strengthen the case for developing biogenic magnetoparticles for multimodal nanomedicine in cancer therapy.


Asunto(s)
Magnetosomas/química , Magnetospirillum/química , Neoplasias Experimentales/radioterapia , Oligopéptidos , Fármacos Sensibilizantes a Radiaciones , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Oligopéptidos/química , Oligopéptidos/farmacología , Terapia de Protones , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Terapia por Rayos X
6.
Int J Nanomedicine ; 14: 7933-7946, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31686819

RESUMEN

BACKGROUND: Human trials combining external radiotherapy (RT) and metallic nanoparticles are currently underway in cancer patients. For internal RT, in which a radioisotope such as radioiodine is systemically administered into patients, there is also a need for enhancing treatment efficacy, decreasing radiation-induced side effects and overcoming radio-resistance. However, if strategies vectorising radioiodine through nanocarriers have been documented, sensitizing the neoplasm through the use of nanotherapeutics easily translatable to the clinic in combination with the standard systemic radioiodine treatment has not been assessed yet. METHOD AND MATERIALS: The present study explored the potential of hybrid poly(methacrylic acid)-grafted gold nanoparticles to improve the performances of systemic 131I-mediated RT on cancer cells and in tumor-bearing mice. Such nanoparticles were chosen based on their ability previously described by our group to safely withstand irradiation doses while exhibiting good biocompatibility and enhanced cellular uptake. RESULTS: In vitro clonogenic assays performed on melanoma and colorectal cancer cells showed that poly(methacrylic acid)-grafted gold nanoparticles (PMAA-AuNPs) could efficiently lead to a marked tumor cell mortality when combined to a low activity of radioiodine, which alone appeared to be essentially ineffective on tumor cells. In vivo, tumor enrichment with PMAA-AuNPs significantly enhanced the killing potential of a systemic radioiodine treatment. CONCLUSION: This is the first report of a simple and reliable nanomedicine-based approach to reduce the dose of radioiodine required to reach curability. In addition, these results open up novel perspectives for using high-Z metallic NPs in additional molecular radiation therapy demonstrating heterogeneous dose distributions.


Asunto(s)
Oro/química , Radioisótopos de Yodo/uso terapéutico , Nanopartículas del Metal/química , Polímeros/química , Animales , Muerte Celular , Línea Celular Tumoral , Femenino , Humanos , Melanoma Experimental/radioterapia , Nanopartículas del Metal/administración & dosificación , Nanopartículas del Metal/ultraestructura , Ratones Endogámicos BALB C , Ratones Desnudos , Ácidos Polimetacrílicos/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Dosificación Radioterapéutica , Simportadores/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
ACS Appl Bio Mater ; 2(1): 144-154, 2019 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-35016337

RESUMEN

In the context of cancer treatment, gold nanoparticles (AuNPs) are considered as very promising radiosensitizers. Here, well-defined polymer-grafted AuNPs were synthesized and studied under gamma irradiation to better understand the involved radiosensitizing mechanisms. First, various water-soluble and well-defined thiol-functionalized homopolymers and copolymers were obtained through atom transfer radical polymerization. They were then used as ligands in the one-step synthesis of AuNPs, which resulted in stable hybrid metal-polymer nanoparticles. Second, these nano-objects were irradiated in solution by γ rays at different doses. Structures were fully characterized through size exclusion chromatography, small-angle X-ray scattering, and small-angle neutron scattering measurements, prior to and after irradiation. We were thus able to quantify and to localize radiation impacts onto the grafted polymers, revealing the production sites of reactive species around AuNPs. Both external and near-surface scissions were observed. Interestingly, the ratio between these two effects was found to vary according to the nature of polymer ligands. Medium-range and long-distance dose enhancements could not be identified from the calculated scission yields, but several mechanisms were considered to explain high yields found for near-surface scissions. Then cytotoxicity was shown to be equivalent for both nonirradiated and irradiated polymer-grafted NPs, which suggested that released polymer fragments were nontoxic. Finally, the potential to add bioactive molecules such as anticancer drugs has been explored by grafting doxorubicin onto the polymer corona. This may lead to nano-objects combining both radiosensitization and chemotherapy effects. This work is the first one to study in details the impact of radiation on radiosensitizing nano-objects combining physical, chemical, and biological analyses.

8.
J Nucl Med ; 59(1): 121-126, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29051343

RESUMEN

Perturbation of thyroid iodide uptake is a well-documented side effect of the use of iodinated contrast media (ICM) administered intravenously. This side effect is thought to be mediated by free iodide in ICM formulations, but this hypothesis has never been formally proven. The aim of the present study was to assess the validity of this hypothesis. Methods: We used mass spectrometry analysis to quantify free-iodide contamination in ICM. Established cell lines expressing the Na/I symporter (NIS) were used to quantify the effect of ICM on iodide uptake. SPECT/CT was used to measure the in vivo uptake of 99mTc-pertechnetate and 123I in 2 NIS-expressing mouse tissues, thyroid and salivary glands. Scintiscans of ICM-naïve and ICM-administered patients were compared. Immunohistologic and Western blot analyses were performed to evaluate NIS protein expression in these organs. Results: Although free iodide was present in ICM formulations, in vitro uptake of iodide by NIS-expressing cells was not significantly affected by ICM. In mice, intravenous or sublingual administration of ICM led to a reduction in radiotracer uptake by the thyroid, accompanied by a dramatic reduction in NIS protein expression in this tissue. In the salivary glands, neither radiotracer uptake nor NIS protein expression was affected by ICM. The thyroid-selective effect of ICM was also observed in humans. Administration of potassium iodide as a source of free iodide led to a diminution of 99mTc-pertechnetate uptake in both mouse thyroid and mouse salivary glands. Altogether, these data rule out a direct intervention of free iodide in the perturbation of thyroid uptake and suggest a direct and selective effect of ICM on the thyroid. Conclusion: We demonstrated that ICM reduce thyroid uptake of iodide independently of free iodide. This effect is due to a specific and dramatic decrease in NIS expression in thyrocytes. These data cast serious doubt on the relevance of measuring urinary iodide concentration to evaluate the delay between ICM administration and radioiodine therapy in patients with differentiated thyroid carcinoma. Finally, the ability of ICM to perturb iodide uptake in the thyroid may be used in radioprotection.


Asunto(s)
Medios de Contraste/química , Medios de Contraste/farmacología , Halogenación , Yoduros/metabolismo , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Células HT29 , Humanos , Ratones , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único , Glándula Tiroides/diagnóstico por imagen
9.
Thyroid ; 26(11): 1614-1622, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27349131

RESUMEN

BACKGROUND: MicroSPECT/CT imaging was used to quantitatively evaluate how iodide uptake in the mouse thyroid is influenced by (i) route of iodine administration; (ii) injection of recombinant human thyrotropin (rhTSH); and (iii) low iodide diet (LID) in euthyroid and triiodothyronine (T3)-treated mice. METHODS: Pertechnetate (99mTcO4-) and 123I thyroid uptake in euthyroid and T3-treated animals fed either a normal-iodine diet (NID) or an LID, treated or not with rhTSH, and radiotracer administered intravenously, subcutaneously, intraperitoneally or by gavage, were assessed using microSPECT/CT imaging. Western blotting was performed to measure sodium/iodide symporter expression levels in the thyroid. RESULTS: Systemic administration of radioiodide resulted in a higher (2.35-fold in NID mice) accumulation of iodide in the thyroid than oral administration. Mice fed LID with systemic radioiodide administration showed a further two-fold increase in thyroid iodide uptake to yield a ∼5-fold increase in uptake compared to the standard NID/oral route. Although rhTSH injections stimulated thyroid activity in both euthyroid and T3-treated mice fed the NID, uptake levels for T3-treated mice remained low compared with those for the euthyroid mice. Combining LID and rhTSH in T3-treated mice resulted in a 2.8-fold higher uptake compared with NID/T3/rhTSH mice and helped restore thyroid activity to levels equivalent to those of euthyroid animals. CONCLUSIONS: Systemic radioiodide administration results in higher thyroidal iodide levels than oral administration, particularly in LID-fed mice. These data highlight the importance of LID, both in euthyroid and T3-treated, rhTSH-injected mice. Extrapolated to human patients, and in the context of clinical guidelines for the preparation of differentiated thyroid cancer patients, our data indicate that LID can potentiate the efficacy of rhTSH treatment in T3-treated patients.


Asunto(s)
Radioisótopos de Yodo/farmacocinética , Radiofármacos/farmacocinética , Glándula Tiroides/diagnóstico por imagen , Triyodotironina/farmacocinética , Administración Oral , Animales , Dieta/efectos adversos , Femenino , Terapia de Reemplazo de Hormonas/efectos adversos , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Inyecciones Subcutáneas , Yodo/administración & dosificación , Yodo/efectos adversos , Radioisótopos de Yodo/administración & dosificación , Radioisótopos de Yodo/metabolismo , Ratones Endogámicos C57BL , Radiofármacos/administración & dosificación , Radiofármacos/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/farmacología , Pertecnetato de Sodio Tc 99m/metabolismo , Pertecnetato de Sodio Tc 99m/farmacocinética , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/metabolismo , Tirotropina/administración & dosificación , Tirotropina/efectos adversos , Tirotropina/farmacología , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único , Triyodotironina/administración & dosificación , Triyodotironina/metabolismo
10.
Biomaterials ; 45: 10-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25662490

RESUMEN

In the present study, we evaluated, in mice, the efficacy of the tetrafunctional block copolymer 704 as a nonviral gene delivery vector to the lungs. SPECT/CT molecular imaging of gene expression, biochemical assays, and immunohistochemistry were used. Our dataset shows that the formulation 704 resulted in higher levels of reporter gene expression than the GL67A formulation currently being used in a clinical trial in cystic fibrosis patients. The inflammatory response associated with this gene transfer was lower than that induced by the GL67A formulation, and the 704 formulation was amenable to repeated administrations. The cell types transfected by the 704 formulation were type I and type II pneumocytes, and transgene expression could not be detected in macrophages. These results emphasize the relevance of the 704 formulation as a nonviral gene delivery vector for lung gene therapy. Further studies will be required to validate this vector in larger animals, in which the lungs are more similar to human lungs.


Asunto(s)
Técnicas de Transferencia de Gen , Pulmón/metabolismo , Polímeros/química , Animales , Cloranfenicol O-Acetiltransferasa/metabolismo , Femenino , Humanos , Inmunohistoquímica , Inflamación/patología , Pulmón/diagnóstico por imagen , Pulmón/patología , Ratones Endogámicos BALB C , Simportadores/metabolismo , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X , Transfección , Transgenes
11.
PLoS One ; 9(3): e92729, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24663284

RESUMEN

Low-energy Auger and conversion electrons deposit their energy in a very small volume (a few nm3) around the site of emission. From a radiotoxicological point of view the effects of low-energy electrons on normal tissues are largely unknown, understudied, and generally assumed to be negligible. In this context, the discovery that the low-energy electron emitter, 99mTc, can induce stunning on primary thyrocytes in vitro, at low absorbed doses, is intriguing. Extrapolated in vivo, this observation suggests that a radioisotope as commonly used in nuclear medicine as 99mTc may significantly influence thyroid physiology. The aims of this study were to determine whether 99mTc pertechnetate (99mTcO4-) is capable of inducing thyroid stunning in vivo, to evaluate the absorbed dose of 99mTcO4- required to induce this stunning, and to analyze the biological events associated/concomitant with this effect. Our results show that 99mTcO4--mediated thyroid stunning can be observed in vivo in mouse thyroid. The threshold of the absorbed dose in the thyroid required to obtain a significant stunning effect is in the range of 20 Gy. This effect is associated with a reduced level of functional Na/I symporter (NIS) protein, with no significant cell death. It is reversible within a few days. At the cellular and molecular levels, a decrease in NIS mRNA, the generation of double-strand DNA breaks, and the activation of the p53 pathway are observed. Low-energy electrons emitted by 99mTc can, therefore, induce thyroid stunning in vivo in mice, if it is exposed to an absorbed dose of at least 20 Gy, a level unlikely to be encountered in clinical practice. Nevertheless this report presents an unexpected effect of low-energy electrons on a normal tissue in vivo, and provides a unique experimental setup to understand the fine molecular mechanisms involved in their biological effects.


Asunto(s)
Pertecnetato de Sodio Tc 99m/efectos adversos , Glándula Tiroides/efectos de la radiación , Animales , Transporte Biológico , Roturas del ADN de Doble Cadena/efectos de la radiación , Electrones , Femenino , Regulación de la Expresión Génica/efectos de la radiación , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Radiometría , Pertecnetato de Sodio Tc 99m/metabolismo , Simportadores/genética , Simportadores/metabolismo , Glándula Tiroides/citología , Glándula Tiroides/metabolismo , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X
12.
PLoS One ; 7(3): e34086, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22470517

RESUMEN

The utilisation of the Na/I symporter (NIS) and associated radiotracers as a reporter system for imaging gene expression is now reaching the clinical setting in cancer gene therapy applications. However, a formal assessment of the methodology in terms of normalisation of the data still remains to be performed, particularly in the context of the assessment of activities in individual subjects in longitudinal studies. In this context, we administered to mice a recombinant, replication-incompetent adenovirus encoding rat NIS, or a human colorectal carcinoma cell line (HT29) encoding mouse NIS. We used (99m)Tc pertechnetate as a radiotracer for SPECT/CT imaging to determine the pattern of ectopic NIS expression in longitudinal kinetic studies. Some animals of the cohort were culled and NIS expression was measured by quantitative RT-PCR and immunohistochemistry. The radioactive content of some liver biopsies was also measured ex vivo. Our results show that in longitudinal studies involving datasets taken from individual mice, the presentation of non-normalised data (activity expressed as %ID/g or %ID/cc) leads to 'noisy', and sometimes incoherent, results. This variability is due to the fact that the blood pertechnetate concentration can vary up to three-fold from day to day. Normalisation of these data with blood activities corrects for these inconsistencies. We advocate that, blood pertechnetate activity should be determined and used to normalise the activity measured in the organ/region of interest that expresses NIS ectopically. Considering that NIS imaging has already reached the clinical setting in the context of cancer gene therapy, this normalisation may be essential in order to obtain accurate and predictive information in future longitudinal clinical studies in biotherapy.


Asunto(s)
Simportadores/análisis , Tomografía Computarizada de Emisión de Fotón Único , Adenoviridae/genética , Animales , Técnicas de Transferencia de Gen , Genes Reporteros , Células HT29 , Humanos , Inmunohistoquímica , Cinética , Hígado/metabolismo , Hígado/patología , Estudios Longitudinales , Ratones , Ratones Endogámicos BALB C , ARN/metabolismo , ARN/normas , Radiofármacos/sangre , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Pertecnetato de Sodio Tc 99m/sangre , Simportadores/genética , Simportadores/metabolismo
13.
PLoS One ; 6(12): e28842, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22205974

RESUMEN

Increased CCL5 levels are markers of an unfavourable outcome in patients with melanoma, breast, cervical, prostate, gastric or pancreatic cancer. Here, we have assessed the role played by CCL5/CCR5 interactions in the development of colon cancer. To do so, we have examined a number of human colorectal carcinoma clinical specimens and found CCL5 and its receptors over-expressed within primary as well as liver and pulmonary metastases of patients compared to healthy tissues. In vitro, CCL5 increased the growth and migratory responses of colon cancer cells from both human and mouse origins. In addition, systemic treatment of mice with CCL5-directed antibodies reduced the extent of development of subcutaneous colon tumors, of liver metastases and of peritoneal carcinosis. Consistently, we found increased numbers of CD45-immunoreactive cells within the stroma of the remaining lesions as well as at the interface with the healthy tissue. In contrast, selective targeting of CCR5 through administration of TAK-779, a CCR5 antagonist, only partially compromised colon cancer progression. Furthermore, CCL5 neutralization rendered the tumors more sensitive to a PDGFRß-directed strategy in mice, this combination regimen offering the greatest protection against liver metastases and suppressing macroscopic peritoneal carcinosis. Collectively, our data demonstrate the involvement of CCL5 in the pathogenesis of colorectal carcinoma and point to its potential value as a therapeutic target.


Asunto(s)
Quimiocina CCL5/antagonistas & inhibidores , Quimiocina CCL5/inmunología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Terapia Molecular Dirigida , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Amidas/farmacología , Animales , Anticuerpos Neutralizantes/inmunología , Células CHO , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimiocina CCL5/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Cricetinae , Cricetulus , Progresión de la Enfermedad , Femenino , Células HT29 , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Ratones , Metástasis de la Neoplasia , Compuestos de Amonio Cuaternario/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptores CCR5/metabolismo , Resultado del Tratamiento
14.
Expert Opin Biol Ther ; 11(10): 1273-85, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21635148

RESUMEN

INTRODUCTION: Progress with gene-based therapies has been hampered by difficulties in monitoring the biodistribution and kinetics of vector-mediated gene expression. Recent developments in non-invasive imaging have allowed researchers and clinicians to assess the location, magnitude and persistence of gene expression in animals and humans. Such advances should eventually lead to improvement in the efficacy and safety of current clinical protocols for future treatments. AREAS COVERED: The molecular imaging techniques for monitoring gene therapy in the living subject, with a specific highlight on the key reporter gene approaches that have been developed and validated in preclinical models using the latest imaging modalities. The applications of molecular imaging to biotherapy, with a particular emphasis on monitoring of gene and vector biodistribution and on image-guided radiotherapy. EXPERT OPINION: Among the reporter gene/probe combinations that have been described so far, one stands out, in our view, as the most versatile and easy to implement: the Na/I symporter. This strategy, exploiting more than 50 years of experience in the treatment of differentiated thyroid carcinomas, has been validated in different types of experimental cancers and with different types of oncolytic viruses and is likely to become a key tool in the implementation of human gene therapy.


Asunto(s)
Expresión Génica , Terapia Genética/métodos , Imagen Molecular/métodos , Trazadores Radiactivos , Animales , Ensayos Clínicos como Asunto/métodos , Ensayos Clínicos como Asunto/tendencias , Diagnóstico por Imagen/métodos , Diagnóstico por Imagen/tendencias , Terapia Genética/tendencias , Humanos , Imagen Molecular/tendencias , Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/tendencias , Tomografía Computarizada de Emisión de Fotón Único/métodos , Tomografía Computarizada de Emisión de Fotón Único/tendencias
15.
Int J Cancer ; 123(2): 365-371, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18404672

RESUMEN

Potassium channels, the most diverse superfamily of ion channels, have recently emerged as regulators of carcinogenesis, thus introducing possible new therapeutic strategies in the fight against cancer. In particular, the large conductance Ca(2+)-activated K(+) channels, often referred to as BK channels, are at the crossroads of several tumor-associated processes such as cell proliferation, survival, secretion and migration. Despite the high BK channel expression in osteosarcoma (OS), their function has not yet been investigated in this malignant bone pathology. Here, using stable RNA interference to reduce the expression of hSlo, the human pore-forming alpha-subunit of the BK channel, in human Cal72 OS cells, we show that BK channels play a functional role in carcinogenesis. Our results reveal for the first time that BK channels exhibit antitumoral properties in OS in vivo and affect the tumor microenvironment through the modulation of both chemokine expression and leukocyte infiltration.


Asunto(s)
Neoplasias Óseas/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Osteosarcoma/metabolismo , Northern Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Plásmidos , Reacción en Cadena de la Polimerasa , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 26(6): 1391-6, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16528004

RESUMEN

OBJECTIVE: Plasma fibronectin (FN) is decreased in several clinical conditions. We were interested to study the thrombotic and hemostatic consequences of the decrease in plasma FN (pFN), the role of FN splice variants in thrombosis, and to examine whether pFN incorporates into thrombi in vivo. METHODS AND RESULTS: We compared the thrombotic response to a vessel injury in FN heterozygous (FN+/-) mice and corresponding FN+/+ mice. Although normal thrombosis in venules was observed, a decrease to half in the pFN concentration in FN+/- mice caused a delay in the appearance of thrombi in arterioles and consequently a delay in their occlusion. We were able to rescue the thrombotic defect in the FN+/- mice by infusion of rat pFN. Additionally, we could show intense incorporation of fluorescent pFN-coated microspheres into the developing thrombi. Moreover, we found that mice expressing FN without the EIIIA or EIIIB domains specific to cellular FN including platelet FN had no thrombotic defect. CONCLUSIONS: Mice heterozygous for FN have a striking defect in thrombus initiation and growth in arterioles attributable to the decrease of pFN. Our study is an example of haploid insufficiency for FN, and it emphasizes the fundamental role of this plasma protein in thrombosis in the arterial system.


Asunto(s)
Arteriolas/efectos de los fármacos , Fibronectinas/sangre , Trombosis/prevención & control , Empalme Alternativo , Animales , Arteriopatías Oclusivas/inducido químicamente , Arteriopatías Oclusivas/complicaciones , Arteriopatías Oclusivas/prevención & control , Tiempo de Sangría , Coagulación Sanguínea/fisiología , Plaquetas/metabolismo , Cloruros , Compuestos Férricos/farmacología , Fibronectinas/genética , Fibronectinas/farmacología , Fibronectinas/fisiología , Haploidia , Heterocigoto , Ratones , Ratones Noqueados , Adhesividad Plaquetaria/fisiología , Estructura Terciaria de Proteína/genética , Ratas , Circulación Esplácnica , Trombosis/inducido químicamente , Trombosis/genética , Trombosis/metabolismo , Vénulas
17.
Trends Mol Med ; 10(4): 179-86, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15059609

RESUMEN

The adhesion receptor P-selectin has long been known to support leukocyte rolling and emigration at sites of inflammation. Recently, P-selectin was also revealed to be a key molecule in hemostasis and thrombosis, mediating platelet rolling, generating procoagulant microparticles containing active tissue factor and enhancing fibrin deposition. Elevated levels of plasma P-selectin are indicative of thrombotic disorders and predictive of future cardiovascular events. Because the interaction between P-selectin and its receptor P-selectin glycoprotein ligand-1 (PSGL-1) represents an important mechanism by which P-selectin induces the formation of procoagulant microparticles and recruits the microparticles to thrombi, anti-thrombotic strategies are currently aimed at inhibiting this interaction. Recent developments also suggest that the procoagulant potential of P-selectin could be used to treat coagulation disorders such as hemophilia A.


Asunto(s)
Selectina-P/fisiología , Animales , Trastornos de la Coagulación Sanguínea/patología , Trastornos de la Coagulación Sanguínea/terapia , Enfermedades Cardiovasculares/patología , Adhesión Celular , Fibrina/metabolismo , Humanos , Inflamación , Leucocitos/metabolismo , Glicoproteínas de Membrana/fisiología , Ratones , Modelos Biológicos , Selectina-P/metabolismo , Fenotipo , Unión Proteica , Estructura Terciaria de Proteína , Tromboplastina/metabolismo , Trombosis/patología
18.
J Clin Invest ; 112(10): 1589-96, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14617760

RESUMEN

Basic and clinical observations suggest that thrombosis and inflammation are closely related. Here we addressed the role played by TNF-alpha in thrombus formation and growth in an in vivo mouse model. Using intravital microscopy, we show that systemic administration of TNF-alpha at doses found in sepsis transiently inhibits thrombus formation and delays arterial occlusion upon vascular injury. These results were reflected in a prolonged bleeding time. Platelets isolated from the TNF-alpha-treated mice showed a marked decrease in fibrinogen binding and P-selectin expression as well as reduced platelet aggregation in response to various agonists. In contrast, in vitro treatment of platelets with TNF-alpha did not affect their function. TNF receptor 1- and 2-deficient mice exhibited normal thrombogenesis in the presence of TNF-alpha. Additionally, the inhibitory effect of TNF-alpha was lost either after treatment with NG-monomethyl-l-arginine, an inhibitor of NO production, or in mice deficient for iNOS. These results indicate that under inflammatory conditions, when leukocytes need free passage to transmigrate into tissues, TNF-alpha decreases platelet activation and inhibits thrombi formation. This effect is not exerted directly on platelets but mediated through the rapid generation of NO in the vessel wall.


Asunto(s)
Fibrinolíticos/uso terapéutico , Trombosis/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , Arterias/anatomía & histología , Arterias/efectos de los fármacos , Arterias/metabolismo , Arterias/patología , Coagulación Sanguínea/fisiología , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Fibrinolíticos/metabolismo , Fibrinolíticos/farmacología , Hemodinámica/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía/métodos , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Selectina-P/genética , Selectina-P/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Trombosis/sangre , Trombosis/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , omega-N-Metilarginina/farmacología
19.
Cell ; 115(2): 139-50, 2003 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-14567912

RESUMEN

Nitric oxide (NO) inhibits vascular inflammation, but the molecular basis for its anti-inflammatory properties is unknown. We show that NO inhibits exocytosis of Weibel-Palade bodies, endothelial granules that mediate vascular inflammation and thrombosis, by regulating the activity of N-ethylmaleimide-sensitive factor (NSF). NO inhibits NSF disassembly of soluble NSF attachment protein receptor (SNARE) complexes by nitrosylating critical cysteine residues of NSF. NO may regulate exocytosis in a variety of physiological processes, including vascular inflammation, neurotransmission, thrombosis, and cytotoxic T lymphocyte cell killing.


Asunto(s)
Proteínas Portadoras/metabolismo , Exocitosis/fisiología , Óxido Nítrico/farmacología , Proteínas de Transporte Vesicular , Animales , Aorta/citología , Proteínas Portadoras/química , Proteínas Portadoras/genética , Células Cultivadas , Cisteína/genética , Cisteína/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Exocitosis/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Proteínas Sensibles a N-Etilmaleimida , NG-Nitroarginina Metil Éster/farmacología , Proteínas Recombinantes/metabolismo , Proteínas SNARE , Trombina/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Cuerpos de Weibel-Palade/efectos de los fármacos , Cuerpos de Weibel-Palade/metabolismo , Factor de von Willebrand/metabolismo
20.
Nat Med ; 9(8): 1020-5, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12858167

RESUMEN

High plasma levels of soluble P-selectin are associated with thrombotic disorders and may predict future cardiovascular events. Mice with high levels of soluble P-selectin have more microparticles in their plasma than do normal mice. Here we show that chimeras of P-selectin and immunoglobulin (P-sel-Ig) induced formation of procoagulant microparticles in human blood through P-selectin glycoprotein ligand-1 (PSGL-1; encoded by the Psgl1 gene, officially known as Selpl). In addition, Psgl1-/- mice produced fewer microparticles after P-sel-Ig infusion and did not spontaneously increase their microparticle count in old age as do wild-type mice. Injected microparticles specifically bound to thrombi and thus could be involved in thrombin generation at sites of injury. Infusion of P-sel-Ig into hemophilia A mice produced a 20-fold increase over control immunoglobulin in microparticles containing tissue factor. This significantly improved the kinetics of fibrin formation in the hemophilia A mice and normalized their tail-bleeding time. P-sel-Ig treatment could become a new approach to sustained control of bleeding in hemophilia.


Asunto(s)
Hemofilia A/terapia , Hemostasis/fisiología , Glicoproteínas de Membrana/metabolismo , Selectina-P/metabolismo , Adulto , Animales , Coagulación Sanguínea/fisiología , Modelos Animales de Enfermedad , Factor VIII/metabolismo , Hemofilia A/metabolismo , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Persona de Mediana Edad , Selectina-P/genética , Fenotipo , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA