Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-29760132

RESUMEN

Aspergillus fumigatus causes life-threatening lung infections in immunocompromised patients. Mouse models are extensively used in research to assess the in vivo efficacies of antifungals. In recent years, there has been an increasing interest in the use of noninvasive imaging techniques to evaluate experimental infections. However, single imaging modalities have limitations concerning the type of information they can provide. In this study, magnetic resonance imaging and bioluminescence imaging were combined to obtain longitudinal information on the extent of developing lesions and fungal load in a leukopenic mouse model of invasive pulmonary aspergillosis (IPA). This multimodal imaging approach was used to assess changes occurring within lungs of infected mice receiving voriconazole treatment starting at different time points after infection. The results showed that IPA development depends on the inoculum size used to infect animals and that disease can be successfully prevented or treated by initiating intervention during early stages of infection. Furthermore, we demonstrated that a reduction in fungal load is not necessarily associated with the disappearance of lesions on anatomical lung images, especially when antifungal treatment coincides with immune recovery. In conclusion, multimodal imaging allows an investigation of different aspects of disease progression or recovery by providing complementary information on dynamic processes, which are highly useful for assessing the efficacy of (novel) therapeutic compounds in a time- and labor-efficient manner.


Asunto(s)
Antifúngicos/uso terapéutico , Aspergillus fumigatus/efectos de los fármacos , Aspergilosis Pulmonar Invasiva/diagnóstico por imagen , Aspergilosis Pulmonar Invasiva/tratamiento farmacológico , Voriconazol/uso terapéutico , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Leucopenia/inmunología , Mediciones Luminiscentes , Pulmón/microbiología , Pulmón/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos BALB C , Imagen Multimodal/métodos , Resultado del Tratamiento
2.
Cytotherapy ; 19(6): 744-755, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28499585

RESUMEN

BACKGROUND AIMS: Myelodysplastic syndromes (MDS) are a group of clonal stem cell disorders affecting the normal hematopoietic differentiation process and leading to abnormal maturation and differentiation of all blood cell lineages. Treatment options are limited, and there is an unmet medical need for effective therapies for patients with severe cytopenias. METHODS: We demonstrate that multipotent adult progenitor cells (MAPC) improve the function of hematopoietic progenitors derived from human MDS bone marrow (BM) by significantly increasing the frequency of primitive progenitors as well as the number of myeloid colonies. RESULTS: This effect was more pronounced in a non-contact culture, indicating the importance of soluble factors produced by the MAPC cells. Moreover, the cells did not stimulate the growth of the abnormal MDS clone, as shown by fluorescent in situ hybridization analysis on BM cells from patients with a known genetic abnormality. We also demonstrate that MAPC cells can provide stromal support for patient-derived hematopoietic cells. When MAPC cells were intravenously injected into a mouse model of MDS, they migrated to the site of injury and increased the hematopoietic function in diseased mice. DISCUSSION: The preclinical studies undertaken here indicate an initial proof of concept for the use of MAPC cell therapy in patients with MDS-related severe and symptomatic cytopenias and should pave the way for further investigation in clinical trials.


Asunto(s)
Células Madre Multipotentes/trasplante , Síndromes Mielodisplásicos/terapia , Adulto , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Femenino , Hematopoyesis , Humanos , Hibridación Fluorescente in Situ , Ratones Endogámicos C57BL
3.
Cancer Res ; 81(8): 1988-2001, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33687947

RESUMEN

Hepatic fat accumulation is associated with diabetes and hepatocellular carcinoma (HCC). Here, we characterize the metabolic response that high-fat availability elicits in livers before disease development. After a short term on a high-fat diet (HFD), otherwise healthy mice showed elevated hepatic glucose uptake and increased glucose contribution to serine and pyruvate carboxylase activity compared with control diet (CD) mice. This glucose phenotype occurred independently from transcriptional or proteomic programming, which identifies increased peroxisomal and lipid metabolism pathways. HFD-fed mice exhibited increased lactate production when challenged with glucose. Consistently, administration of an oral glucose bolus to healthy individuals revealed a correlation between waist circumference and lactate secretion in a human cohort. In vitro, palmitate exposure stimulated production of reactive oxygen species and subsequent glucose uptake and lactate secretion in hepatocytes and liver cancer cells. Furthermore, HFD enhanced the formation of HCC compared with CD in mice exposed to a hepatic carcinogen. Regardless of the dietary background, all murine tumors showed similar alterations in glucose metabolism to those identified in fat exposed nontransformed mouse livers, however, particular lipid species were elevated in HFD tumor and nontumor-bearing HFD liver tissue. These findings suggest that fat can induce glucose-mediated metabolic changes in nontransformed liver cells similar to those found in HCC. SIGNIFICANCE: With obesity-induced hepatocellular carcinoma on a rising trend, this study shows in normal, nontransformed livers that fat induces glucose metabolism similar to an oncogenic transformation.


Asunto(s)
Carcinoma Hepatocelular/etiología , Dieta Alta en Grasa , Grasas de la Dieta/metabolismo , Glucosa/metabolismo , Hepatocitos/metabolismo , Neoplasias Hepáticas/etiología , Animales , Carcinoma Hepatocelular/metabolismo , Transformación Celular Neoplásica , Ciclo del Ácido Cítrico/fisiología , Ácidos Grasos/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Ácido Láctico/biosíntesis , Metabolismo de los Lípidos , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones , Palmitatos/farmacología , Peroxisomas/metabolismo , Proteómica , Piruvato Carboxilasa/metabolismo , Distribución Aleatoria , Especies Reactivas de Oxígeno/metabolismo , Serina/metabolismo , Activación Transcripcional
4.
Mol Ther Methods Clin Dev ; 17: 317-327, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32577429

RESUMEN

Stem cell-based therapies are a promising approach for the treatment of degenerative muscular diseases; however, clinical trials have shown inconclusive and even disappointing results so far. Noninvasive cell monitoring by medicine imaging could improve the understanding of the survival and biodistribution of cells following injection. In this study, we assessed the canine sodium iodide symporter (cNIS) reporter gene as an imaging tool to track by single-photon emission computed tomography (SPECT/CT) transduced canine myoblasts after intramuscular (IM) administrations in dogs. cNIS-expressing cells kept their myogenic capacities and showed strong 99 mTc-pertechnetate (99 mTcO4 -) uptake efficiency both in vitro and in vivo. cNIS expression allowed visualization of cells by SPECT/CT along time: 4 h, 48 h, 7 days, and 30 days after IM injection; biopsies collected 30 days post administration showed myofiber's membranes expressing cNIS. This study demonstrates that NIS can be used as a reporter to track cells in vivo in the skeletal muscle of large animals. Our results set a proof of concept of the benefits NIS-tracking tool may bring to the already challenging cell-based therapies arena in myopathies and pave the way to a more efficient translation to the clinical setting from more accurate pre-clinical results.

5.
Methods Mol Biol ; 1895: 123-134, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30539534

RESUMEN

Cells expressing suicide genes can be used as therapeutic vehicles for difficult-to-treat tumors, for example, if stem cells are used that are able to track infiltrating tumor cells. An alternative application of suicide gene expression is their use as a safety switch in regenerative medicine where the presence of a few pluripotent stem cells could potentially cause unwanted side effects like the formation of teratoma. One potential bottleneck of these applications is that information on the initiation of cell suicide is needed early on, for example, when therapeutic cells have reached infiltrating tumor cells or when teratomas are formed. Therefore, in vivo imaging methods are needed that provide information on target location, (stem) cell location, (stem) cell viability, pathology, and suicide gene expression. This requires multimodal imaging approaches that can provide this information longitudinally and in a noninvasive way. Here, we describe examples of how therapeutic cells can be modified so that they express a suicide gene and genes that can be used for in vivo visualization.


Asunto(s)
Genes Transgénicos Suicidas , Terapia Genética/métodos , Células Madre Embrionarias Humanas , Imagen Molecular/métodos , Imagen Multimodal/métodos , Neoplasias/diagnóstico por imagen , Animales , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/terapia , Modelos Animales de Enfermedad , Humanos , Imagen por Resonancia Magnética , Ratones , Ratones Desnudos , Neoplasias/terapia , Tomografía de Emisión de Positrones
6.
Theranostics ; 8(10): 2799-2813, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29774076

RESUMEN

Rationale: Pluripotent stem cells (PSCs) are being investigated as a cell source for regenerative medicine since they provide an infinitive pool of cells that are able to differentiate towards every cell type of the body. One possible therapeutic application involves the use of these cells to treat myocardial infarction (MI), a condition where billions of cardiomyocytes (CMs) are lost. Although several protocols have been developed to differentiate PSCs towards CMs, none of these provide a completely pure population, thereby still posing a risk for neoplastic teratoma formation. Therefore, we developed a strategy to (i) monitor cell behavior noninvasively via site-specific integration of firefly luciferase (Fluc) and the human positron emission tomography (PET) imaging reporter genes, sodium iodide symporter (hNIS) and somatostatin receptor type 2 (hSSTr2), and (ii) perform hSSTr2-mediated suicide gene therapy via the clinically used radiopharmacon 177Lu-DOTATATE. Methods: Human embryonic stem cells (ESCs) were gene-edited via zinc finger nucleases to express Fluc and either hNIS or hSSTr2 in the safe harbor locus, adeno-associated virus integration site 1. Firstly, these cells were exposed to 4.8 MBq 177Lu-DOTATATE in vitro and cell survival was monitored via bioluminescence imaging (BLI). Afterwards, hNIS+ and hSSTr2+ ESCs were transplanted subcutaneously and teratomas were allowed to form. At day 59, baseline 124I and 68Ga-DOTATATE PET and BLI scans were performed. The day after, animals received either saline or 55 MBq 177Lu-DOTATATE. Weekly BLI scans were performed, accompanied by 124I and 68Ga-DOTATATE PET scans at days 87 and 88, respectively. Finally, hSSTr2+ ESCs were differentiated towards CMs and transplanted intramyocardially in the border zone of an infarct that was induced by left anterior descending coronary artery ligation. After transplantation, the animals were monitored via BLI and PET, while global cardiac function was evaluated using cardiac magnetic resonance imaging. Results: Teratoma growth of both hNIS+ and hSSTr2+ ESCs could be followed noninvasively over time by both PET and BLI. After 177Lu-DOTATATE administration, successful cell killing of the hSSTr2+ ESCs was achieved both in vitro and in vivo, indicated by reductions in total tracer lesion uptake, BLI signal and teratoma volume. As undifferentiated hSSTr2+ ESCs are not therapeutically relevant, they were differentiated towards CMs and injected in immune-deficient mice with a MI. Long-term cell survival could be monitored without uncontrolled cell proliferation. However, no improvement in the left ventricular ejection fraction was observed. Conclusion: We developed isogenic hSSTr2-expressing ESCs that allow noninvasive cell monitoring in the context of PSC-derived regenerative therapy. Furthermore, we are the first to use the hSSTr2 not only as an imaging reporter gene, but also as a suicide mechanism for radionuclide therapy in the setting of PSC-derived cell treatment.


Asunto(s)
Células Madre Embrionarias Humanas/trasplante , Infarto del Miocardio/terapia , Receptores de Somatostatina/metabolismo , Trasplante de Células Madre/métodos , Teratoma/diagnóstico por imagen , Animales , Línea Celular , Femenino , Genes Reporteros , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Desnudos , Infarto del Miocardio/diagnóstico por imagen , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Octreótido/análogos & derivados , Compuestos Organometálicos , Tomografía de Emisión de Positrones , Radiofármacos , Receptores de Somatostatina/genética , Trasplante de Células Madre/efectos adversos , Teratoma/etiología
7.
Mol Imaging Biol ; 20(6): 940-951, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29671177

RESUMEN

PURPOSE: Transplantation of pancreatic islets (PIs) is a promising therapeutic approach for type 1 diabetes. The main obstacle for this strategy is that the outcome of islet engraftment depends on the engraftment site. It was our aim to develop a strategy for using non-invasive imaging techniques to assess the location and fate of transplanted PIs longitudinally in vivo. PROCEDURES: In order to overcome the limitations of individual imaging techniques and cross-validate findings by different modalities, we have combined fluorine magnetic resonance imaging (F-19 MRI), fluorescence imaging (FLI), and bioluminescent imaging (BLI) for studying subcutaneously transplanted PIs and beta cell-like cells (INS-1E cell line) in vivo. We optimized the transduction (using lentiviral vectors) and labeling procedures (using perfluoro crown ether nanoparticles with a fluorescence dye) for PIs and INS-1E cell imaging. RESULTS: The feasibility of using the proposed imaging methods for PI assessment was demonstrated both in vitro and in vivo. Our data suggested that F-19 MRI is suitable for high-resolution localization of transplanted cells and PIs; FLI is essential for confirmation of contrast localization by histology; and BLI is a reliable method to assess cell viability and survival after transplantation. No significant side effects on cell viability and function have been observed. CONCLUSIONS: The proposed tri-modal imaging platform is a valuable approach for the assessment of engrafted PIs in vivo. It is potentially suitable for comparing different transplantation sites and evaluating novel strategies for improving PI transplantation technique in the future.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/diagnóstico por imagen , Imagen Multimodal/métodos , Tejido Subcutáneo/diagnóstico por imagen , Animales , Línea Celular , Femenino , Humanos , Ratones , Ratones Desnudos , Ratas Wistar
8.
Nat Commun ; 8(1): 1249, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29093487

RESUMEN

Muscular dystrophies (MDs) are often characterized by impairment of both skeletal and cardiac muscle. Regenerative strategies for both compartments therefore constitute a therapeutic avenue. Mesodermal iPSC-derived progenitors (MiPs) can regenerate both striated muscle types simultaneously in mice. Importantly, MiP myogenic propensity is influenced by somatic lineage retention. However, it is still unknown whether human MiPs have in vivo potential. Furthermore, methods to enhance the intrinsic myogenic properties of MiPs are likely needed, given the scope and need to correct large amounts of muscle in the MDs. Here, we document that human MiPs can successfully engraft into the skeletal muscle and hearts of dystrophic mice. Utilizing non-invasive live imaging and selectively induced apoptosis, we report evidence of striated muscle regeneration in vivo in mice by human MiPs. Finally, combining RNA-seq and miRNA-seq data, we define miRNA cocktails that promote the myogenic potential of human MiPs.


Asunto(s)
Corazón/crecimiento & desarrollo , Células Madre Pluripotentes Inducidas/citología , Mesodermo/citología , MicroARNs/genética , Desarrollo de Músculos/genética , Músculo Esquelético/crecimiento & desarrollo , Distrofia Muscular Animal/patología , Miocardio/citología , Animales , Diferenciación Celular , Ecocardiografía , Corazón/diagnóstico por imagen , Humanos , Ratones , Músculo Esquelético/citología , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/patología , Distrofia Muscular Animal/diagnóstico por imagen , Miocardio/patología , Regeneración
9.
J Nucl Med ; 58(10): 1659-1665, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28596158

RESUMEN

Molecular imaging is indispensable for determining the fate and persistence of engrafted stem cells. Standard strategies for transgene induction involve the use of viral vectors prone to silencing and insertional mutagenesis or the use of nonhuman genes. Methods: We used zinc finger nucleases to induce stable expression of human imaging reporter genes into the safe-harbor locus adeno-associated virus integration site 1 in human embryonic stem cells. Plasmids were generated carrying reporter genes for fluorescence, bioluminescence imaging, and human PET reporter genes. Results: In vitro assays confirmed their functionality, and embryonic stem cells retained differentiation capacity. Teratoma formation assays were performed, and tumors were imaged over time with PET and bioluminescence imaging. Conclusion: This study demonstrates the application of genome editing for targeted integration of human imaging reporter genes in human embryonic stem cells for long-term molecular imaging.


Asunto(s)
Células Madre Embrionarias/metabolismo , Endorribonucleasas/metabolismo , Edición Génica , Genes Reporteros/genética , Genoma Humano/genética , Tomografía de Emisión de Positrones , Dedos de Zinc , Animales , Diferenciación Celular , Línea Celular , Endorribonucleasas/química , Femenino , Expresión Génica , Humanos , Hígado/citología , Ratones
10.
Stem Cells Int ; 2016: 6235687, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26997958

RESUMEN

Numerous neurodegenerative and neuromuscular disorders are associated with cell-specific depletion in the human body. This imbalance in tissue homeostasis is in healthy individuals repaired by the presence of endogenous stem cells that can replace the lost cell type. However, in most disorders, a genetic origin or limited presence or exhaustion of stem cells impairs correct cell replacement. During the last 30 years, methods to readily isolate and expand stem cells have been developed and this resulted in a major change in the regenerative medicine field as it generates sufficient amount of cells for human transplantation applications. Furthermore, stem cells have been shown to release cytokines with beneficial effects for several diseases. At present however, clinical stem cell transplantations studies are struggling to demonstrate clinical efficacy despite promising preclinical results. Therefore, to allow stem cell therapy to achieve its full potential, more insight in their in vivo behavior has to be achieved. Different methods to noninvasively monitor these cells have been developed and are discussed. In some cases, stem cell monitoring even reached the clinical setting. We anticipate that by further exploring these imaging possibilities and unraveling their in vivo behavior further improvement in stem cell transplantations will be achieved.

11.
Stem Cell Reports ; 5(6): 1183-1195, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26626179

RESUMEN

Muscular dystrophies are a heterogeneous group of myopathies, characterized by muscle weakness and degeneration, without curative treatment. Mesoangioblasts (MABs) have been proposed as a potential regenerative therapy. To improve our understanding of the in vivo behavior of MABs and the effect of different immunosuppressive therapies, like cyclosporine A or co-stimulation-adhesion blockade therapy, on cell survival noninvasive cell monitoring is required. Therefore, cells were transduced with a lentiviral vector encoding firefly luciferase (Fluc) and the human sodium iodide transporter (hNIS) to allow cell monitoring via bioluminescence imaging (BLI) and small-animal positron emission tomography (PET). Non-H2 matched mMABs were injected in the femoral artery of dystrophic mice and were clearly visible via small-animal PET and BLI. Based on noninvasive imaging data, we were able to show that co-stim was clearly superior to CsA in reducing cell rejection and this was mediated via a reduction in cytotoxic T cells and upregulation of regulatory T cells.


Asunto(s)
Vasos Sanguíneos/citología , Desarrollo de Músculos , Distrofia Muscular Animal/terapia , Tomografía de Emisión de Positrones/métodos , Trasplante de Células Madre , Células Madre/citología , Simportadores/análisis , Animales , Línea Celular , Supervivencia Celular , Ciclosporina/uso terapéutico , Genes Reporteros , Humanos , Inmunosupresores/uso terapéutico , Luciferasas de Luciérnaga/análisis , Luciferasas de Luciérnaga/genética , Mediciones Luminiscentes , Ratones Endogámicos C57BL , Ratones Desnudos , Distrofia Muscular Animal/diagnóstico , Distrofia Muscular Animal/patología , Imagen Óptica , Simportadores/genética , Transducción Genética
12.
PLoS One ; 9(4): e94833, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24747914

RESUMEN

PURPOSE: The use of stably integrated reporter gene imaging provides a manner to monitor the in vivo fate of engrafted cells over time in a non-invasive manner. Here, we optimized multimodal imaging (small-animal PET, Cerenkov luminescence imaging (CLI) and bioluminescence imaging (BLI)) of mesenchymal stem cells (MSCs), by means of the human sodium iodide symporter (hNIS) and firefly luciferase (Fluc) as reporters. METHODS: First, two multicistronic lentiviral vectors (LV) were generated for multimodal imaging: BLI, 124I PET/SPECT and CLI. Expression of the imaging reporter genes was validated in vitro using 99mTcO4- radioligand uptake experiments and BLI. Uptake kinetics, specificity and tracer elution were determined as well as the effect of the transduction process on the cell's differentiation capacity. MSCs expressing the LV were injected intravenously or subcutaneously and imaged using small-animal PET, CLI and BLI. RESULTS: The expression of both imaging reporter genes was functional and specific. An elution of 99mTcO4- from the cells was observed, with 31% retention after 3 h. After labeling cells with 124I in vitro, a significantly higher CLI signal was noted in hNIS expressing murine MSCs. Furthermore, it was possible to visualize cells injected intravenously using BLI or subcutaneously in mice, using 124I small-animal PET, CLI and BLI. CONCLUSIONS: This study identifies hNIS as a suitable reporter gene for molecular imaging with PET and CLI, as confirmed with BLI through the expression of Fluc. It supports the potential for a wider application of hNIS reporter gene imaging and future clinical applications.


Asunto(s)
Luminiscencia , Células Madre Mesenquimatosas/metabolismo , Imagen Multimodal/métodos , Imagen Óptica/métodos , Tomografía de Emisión de Positrones/métodos , Simportadores/genética , Animales , Diferenciación Celular/efectos de los fármacos , Genes Reporteros/genética , Vectores Genéticos/genética , Células HEK293 , Humanos , Radioisótopos de Yodo , Lentivirus/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Factor 1 de Elongación Peptídica/genética , Puromicina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA