Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Youth Adolesc ; 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38782845

RESUMEN

Although smartphone ownership among minors has become an important social phenomenon, its impact on children's and adolescents' well-being, as well as the mechanisms by which this might take place are not yet sufficiently well-established. To date, no research has examined the effect of smartphone ownership on the well-being of minors through the consumption of influencer-generated content, nor has it explored the effectiveness of the main prevention strategies employed by parents in this context. To fill those gaps, 800 Spanish minors (50% female) aged from 8 to 16 years old (M = 12.33, SD = 2.38) participated in a correlational study in which the ownership of electronic devices, the consumption of influencer generated content, the parasocial relationship with the influencer, and the most common parental mediation strategies were considered. The results showed a positive association between electronic device ownership and psychological discomfort, problematic usage, and imitation of dangerous behaviors. This association was mediated by the consumption of influencer-generated content and the parasocial relationship established by the minor with the influencer. Regarding preventive strategies, only active mediation was inversely related to poorer well-being indicators, however this positive effect significantly decreased when a smartphone or a similar electronic device was owned by the minor (vs. no owned). These findings contribute to the understanding of how smartphone ownership can affect the well-being of children, emphasizing the need for thoughtful consideration when deciding whether to provide smartphones to minors.

2.
Cancer Immunol Immunother ; 72(8): 2623-2633, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37062034

RESUMEN

Immunotherapy with chimeric antigen receptor T (CAR T) cells has changed the treatment of hematological malignances, but they are still a challenge for solid tumors, including pediatric sarcomas. Here, we report a switchable CAR T cell strategy based on anti-FITC CAR T cells and a switch molecule conjugated with FITC for targeting osteosarcoma (OS) tumors. As a potential target, we analyzed the expression of B7-H3, an immune checkpoint inhibitor, in OS cell lines. In addition, we evaluate the capacity of an anti-B7-H3 monoclonal antibody conjugated with FITC (anti-B7-H3-FITC mAb) to control the antitumor activity of anti-FITC CAR T cells. The effector functions of anti-FITC CAR T cells against OS, measured in vitro by tumor cell killing activity and cytokine production, are dependent on the presence of the anti-B7-H3-FITC mAb switch. Moreover, OS cells stimulate anti-FITC CAR T cells migration. In vivo, anti-B7-H3 mAb penetrates in the tumor and binds 143B OS tumor cells. Furthermore, anti-FITC CAR T cells reach tumor region and exert antitumor effect in an OS NSG mouse model only in the presence of the switch molecule. We demonstrate that anti-B7-H3-FITC mAb redirects the cytotoxic activity of anti-FITC CAR T cells against OS tumors suggesting that switchable CAR T cell platforms might be a plausible strategy against OS.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Niño , Linfocitos T , Fluoresceína-5-Isotiocianato/metabolismo , Antígenos B7/metabolismo , Osteosarcoma/terapia , Anticuerpos Monoclonales , Neoplasias Óseas/terapia , Línea Celular Tumoral , Inmunoterapia Adoptiva
3.
Curr Psychol ; : 1-12, 2022 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-36373116

RESUMEN

Increasing incidence of problem gambling has led to prioritizing the problem from the point of view of public health. Additionally, gambling disorder has been recently classified as a behavioral addiction, with implications for both its diagnosis and treatment. However, the shared neural substrate of addictions, to substances and behavioral, is still discussed. Thus, this systematic review aims to provide up-to-date knowledge from the past five years (2017-2022) concerning the neural correlates of gambling related stimuli (cue-reactivity) on the basis of a previous review (Brevers et al., Cognitive, Affective and Behavioral Neuroscience 18:718-729, 2019). A total of five studies were included in the review. Activation of brain areas related to memory, reward and executive functions could be the underlying mechanism of this behavioral addiction. Specifically, nucleus accumbens and striatum (ventral and dorsal), parahippocampal regions, the right amygdala and several prefrontal cortex regions have systematically been found more active in those subjects exposed to gambling-related cues. Also, the insula could play a pivotal role connecting these three systems in a highly integrated neural network with several implications for reward processing modulation, associative learning and top-down attentional regulation to improve saliency of addiction-related cues. These results are consistent with previous findings on other substance addictions, such as alcohol, tobacco, marijuana or cocaine. The study of neural reactivity to stimuli related to addiction could be useful as a biomarker of the severity of the disorder, the efficacy of the treatment, the risk of relapse, in addition to being an objective criterion to measure the effectiveness of prevention campaigns.

4.
Vet Res ; 52(1): 22, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33588935

RESUMEN

COVID-19 is a zoonotic disease caused by SARS-CoV-2. Infections of animals with SARS-CoV-2 have recently been reported, and an increase of severe lung pathologies in domestic dogs has also been detected by veterinarians in Spain. Therefore, further descriptions of the pathological processes in those animals that show symptoms similar to those described in humans affected by COVID-19 would be highly valuable. The potential for companion animals to contribute to the continued transmission and community spread of this known human-to-human disease is an urgent issue to be considered. Forty animals with pulmonary pathologies were studied by chest X-ray, ultrasound analysis, and computed tomography. Nasopharyngeal and rectal swabs were analyzed to detect canine pathogens, including SARS-CoV-2. An additional twenty healthy dogs living in SARS-CoV-2-positive households were included. Immunoglobulin detection by several immunoassays was performed. Our findings show that sick dogs presented severe alveolar or interstitial patterns with pulmonary opacity, parenchymal abnormalities, and bilateral lesions. The forty sick dogs were negative for SARS-CoV-2 but Mycoplasma spp. was detected in 26 of 33 dogs. Five healthy and one pathological dog presented IgG against SARS-CoV-2. Here we report that despite detecting dogs with α-SARS-CoV-2 IgG, we never obtained a positive RT-qPCR for SARS-SoV-2, not even in dogs with severe pulmonary disease; suggesting that even in the case of canine infection, transmission would be unlikely. Moreover, dogs living in COVID-19-positive households could have been more highly exposed to infection with SARS-CoV-2.


Asunto(s)
COVID-19/veterinaria , Enfermedades de los Perros/transmisión , Inmunoglobulinas/sangre , Zoonosis/transmisión , Animales , COVID-19/transmisión , COVID-19/virología , Enfermedades de los Perros/virología , Perros , Femenino , Inmunidad Humoral , Masculino , España , Zoonosis/virología
5.
Mol Ther ; 28(4): 1033-1042, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32053771

RESUMEN

We present here the results of a first-in-human, first-in-child trial for patients with relapsed/refractory solid tumors using Celyvir, an advanced therapy medicine that combines autologous mesenchymal stem cells (MSCs) carrying an oncolytic adenovirus. Celyvir was manufactured from a bone marrow aspirate and then given intravenously. Patients received weekly infusions for 6 weeks at a dose of 2 × 106 cells/kg (children) or 0.5-1 × 106 cells/kg (adults), 2 × 104 viral particles per cell. Fifteen pediatric and 19 adult patients were recruited, but 18 were screen failures, mainly because rapid disease progression before Celyvir was available. No grade 2-5 toxicities were reported. Adenoviral replication detected by PCR was found in all but 2 pediatric patient and in none of the adult ones. Absolute numbers of circulating leukocytes suffered minor changes along therapy, but some subsets showed differences comparing the pediatric versus the adult cohorts. Two patients with neuroblastoma showed disease stabilization, and one of them continued on treatment for up to 6 additional weeks. Celyvir, the combination of MSCs and oncolytic adenovirus, is safe and warrants further evaluation in a phase 2 setting. The use of MSCs may be a strategy to increase the amount of oncolytic virus administered to patients, minimizing toxicities and avoiding direct tumor injections.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/virología , Neoplasias/terapia , Virus Oncolíticos/genética , Adolescente , Adulto , Factores de Edad , Anciano , Niño , Preescolar , Dependovirus/genética , Dependovirus/fisiología , Estudios de Factibilidad , Humanos , Persona de Mediana Edad , Neoplasias/inmunología , Virus Oncolíticos/fisiología , Trasplante Autólogo , Resultado del Tratamiento
6.
Genome Res ; 27(3): 335-348, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27965292

RESUMEN

Half the human genome is made of transposable elements (TEs), whose ongoing activity continues to impact our genome. LINE-1 (or L1) is an autonomous non-LTR retrotransposon in the human genome, comprising 17% of its genomic mass and containing an average of 80-100 active L1s per average genome that provide a source of inter-individual variation. New LINE-1 insertions are thought to accumulate mostly during human embryogenesis. Surprisingly, the activity of L1s can further impact the somatic human brain genome. However, it is currently unknown whether L1 can retrotranspose in other somatic healthy tissues or if L1 mobilization is restricted to neuronal precursor cells (NPCs) in the human brain. Here, we took advantage of an engineered L1 retrotransposition assay to analyze L1 mobilization rates in human mesenchymal (MSCs) and hematopoietic (HSCs) somatic stem cells. Notably, we have observed that L1 expression and engineered retrotransposition is much lower in both MSCs and HSCs when compared to NPCs. Remarkably, we have further demonstrated for the first time that engineered L1s can retrotranspose efficiently in mature nondividing neuronal cells. Thus, these findings suggest that the degree of somatic mosaicism and the impact of L1 retrotransposition in the human brain is likely much higher than previously thought.


Asunto(s)
Elementos Transponibles de ADN , Elementos de Nucleótido Esparcido Largo , Células-Madre Neurales/metabolismo , División Celular , Células Cultivadas , Células HeLa , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Mosaicismo , Células-Madre Neurales/citología
7.
Stem Cells ; 36(10): 1487-1500, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30001480

RESUMEN

Osteosarcoma (OS) is a highly aggressive bone tumor that usually arises intramedullary at the extremities of long bones. Due to the fact that the peak of incidence is in the growth spurt of adolescence, the specific anatomical location, and the heterogeneity of cells, it is believed that osteosarcomagenesis is a process associated with bone development. Different studies in murine models showed that the tumor-initiating cell in OS could be an uncommitted mesenchymal stem cell (MSC) developing in a specific bone microenvironment. However, only a few studies have reported transgene-induced human MSCs transformation and mostly obtained undifferentiated sarcomas. In our study, we demonstrate that activator protein 1 family members induce osteosarcomagenesis in immortalized hMSC. c-JUN or c-JUN/c-FOS overexpression act as tumorigenic factors generating OS with fibroblastic or pleomorphic osteoblastic phenotypes, respectively. Stem Cells 2018;36:1487-1500.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Osteosarcoma/metabolismo , Osteosarcoma/patología , Factor de Transcripción AP-1/metabolismo , Animales , Xenoinjertos , Humanos , Ratones , Ratones SCID , Fenotipo
8.
Cancer Immunol Immunother ; 67(10): 1589-1602, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30066102

RESUMEN

Oncolytic virotherapy uses oncolytic viruses that selectively replicate in cancer cells. The use of cellular vehicles with migration ability to tumors has been considered to increase their delivery to target sites. Following this approach, the antitumor efficacy of the treatment Celyvir (mesenchymal stem cells infected with the oncolytic adenovirus ICOVIR-5) has been demonstrated in patients with neuroblastoma. However, the better efficacy of syngeneic or allogeneic mesenchymal stem cells as cell carriers and the specific role of the immune system in this therapy are still unknown. In this study we use our virotherapy Celyvir with syngeneic and allogeneic mouse mesenchymal stem cells to determine their antitumor efficacy in a C57BL/6 murine adenocarcinoma model. Adoptive transfer of splenocytes from treated mice to new tumor-bearing mice followed by a secondary adoptive transfer to a third group was performed. Similar reduction of tumor growth and systemic activation of the innate and adaptive immune system was observed in groups treated with syngeneic or allogeneic mesenchymal stem cells loaded with ICOVIR-5. Moreover, a different pattern of infiltration was observed by immunofluorescence in Celyvir-treated groups. While non-treated tumors presented higher density of infiltrating immune cells in the periphery of the tumor, both syngeneic and allogeneic Celyvir-treated groups presented higher infiltration of CD45+ cells in the core of the tumor. Therefore, these results suggest that syngeneic and allogeneic Celyvir induce systemic activation of the immune system, similar antitumor effect and a higher intratumoral infiltration of leukocytes.


Asunto(s)
Adenocarcinoma/terapia , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Linfocitos Infiltrantes de Tumor/inmunología , Células Madre Mesenquimatosas/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/genética , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Femenino , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos C57BL , Células Tumorales Cultivadas
9.
Genome Res ; 25(1): 27-40, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25271306

RESUMEN

In differentiated cells, aging is associated with hypermethylation of DNA regions enriched in repressive histone post-translational modifications. However, the chromatin marks associated with changes in DNA methylation in adult stem cells during lifetime are still largely unknown. Here, DNA methylation profiling of mesenchymal stem cells (MSCs) obtained from individuals aged 2 to 92 yr identified 18,735 hypermethylated and 45,407 hypomethylated CpG sites associated with aging. As in differentiated cells, hypermethylated sequences were enriched in chromatin repressive marks. Most importantly, hypomethylated CpG sites were strongly enriched in the active chromatin mark H3K4me1 in stem and differentiated cells, suggesting this is a cell type-independent chromatin signature of DNA hypomethylation during aging. Analysis of scedasticity showed that interindividual variability of DNA methylation increased during aging in MSCs and differentiated cells, providing a new avenue for the identification of DNA methylation changes over time. DNA methylation profiling of genetically identical individuals showed that both the tendency of DNA methylation changes and scedasticity depended on nongenetic as well as genetic factors. Our results indicate that the dynamics of DNA methylation during aging depend on a complex mixture of factors that include the DNA sequence, cell type, and chromatin context involved and that, depending on the locus, the changes can be modulated by genetic and/or external factors.


Asunto(s)
Envejecimiento/genética , Metilación de ADN , ADN/genética , Células Madre/citología , Adolescente , Anciano , Anciano de 80 o más Años , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Cromatina/genética , Epigénesis Genética , Histonas/genética , Humanos , Análisis por Micromatrices , Persona de Mediana Edad , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Análisis de Secuencia de ADN , Gemelos Monocigóticos , Adulto Joven
11.
Adv Exp Med Biol ; 1031: 371-386, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29214583

RESUMEN

Rare diseases are highly diverse and complex regarding molecular underpinning and clinical manifestation and afflict millions of patients worldwide. The lack of appropriate model systems with face and construct validity and the limited availability of live tissues and cells from patients has largely hampered the understanding of underlying disease mechanisms. As a consequence, there are no adequate treatment options available for the vast majority of rare diseases. Over the last decade, remarkable progress in pluripotent and adult stem cell biology and the advent of powerful genomic technologies opened up exciting new avenues for the investigation, diagnosis, and personalized therapy of intractable human diseases. Utilizing the entire range of available stem cell types will continue to cross-fertilize different research areas and leverage the investigation of rare diseases based on evidence-based medicine. Standardized cell engineering and manufacturing from inexhaustible stem cell sources should lay the foundation for next-generation drug discovery and cell therapies that are broadly applicable in regenerative medicine. In this chapter we discuss how patient- and disease-specific iPS cells as well as adult stem cells are changing the pace of biomedical research and the translational landscape.


Asunto(s)
Células Madre Adultas/trasplante , Células Madre Pluripotentes/trasplante , Enfermedades Raras/cirugía , Medicina Regenerativa/métodos , Proyectos de Investigación , Trasplante de Células Madre/métodos , Ensayos Clínicos como Asunto/métodos , Humanos , Enfermedades Raras/diagnóstico , Enfermedades Raras/epidemiología , Resultado del Tratamiento
12.
J Transl Med ; 14(1): 207, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27393146

RESUMEN

BACKGROUND: Age-associated changes in genomic DNA methylation have been primarily attributed to 5-methylcytosine (5mC). However, the recent discovery of 5-hydroxymethylcytosine (5hmC) suggests that this epigenetic mark might also play a role in the process. METHODS: Here, we analyzed the genome-wide profile of 5hmc in mesenchymal stem cells (MSCs) obtained from bone-marrow donors, aged 2-89 years. RESULTS: We identified 10,685 frequently hydroxymethylated CpG sites in MSCs that were, as in other cell types, significantly associated with low density CpG regions, introns, the histone posttranslational modification H3k4me1 and enhancers. Study of the age-associated changes to 5hmC identified 785 hyper- and 846 hypo-hydroxymethylated CpG sites in the MSCs obtained from older individuals. CONCLUSIONS: DNA hyper-hydroxymethylation in the advanced-age group was associated with loss of 5mC, which suggests that, at specific CpG sites, this epigenetic modification might play a role in DNA methylation changes during lifetime. Since bone-marrow MSCs have many clinical applications, and the fact that the epigenomic alterations in this cell type associated with aging identified in this study could have associated functional effects, the age of donors should be taken into account in clinical settings.


Asunto(s)
5-Metilcitosina/análogos & derivados , Envejecimiento/genética , Células de la Médula Ósea/citología , Metilación de ADN/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , 5-Metilcitosina/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Cromatina/metabolismo , Islas de CpG/genética , Genoma Humano , Genómica , Humanos , Persona de Mediana Edad , Adulto Joven
13.
Cell Mol Life Sci ; 72(16): 3097-113, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25935149

RESUMEN

The bone is a complex connective tissue composed of many different cell types such as osteoblasts, osteoclasts, chondrocytes, mesenchymal stem/progenitor cells, hematopoietic cells and endothelial cells, among others. The interaction between them is finely balanced through the processes of bone formation and bone remodeling, which regulates the production and biological activity of many soluble factors and extracellular matrix components needed to maintain the bone homeostasis in terms of cell proliferation, differentiation and apoptosis. Osteosarcoma (OS) emerges in this complex environment as a result of poorly defined oncogenic events arising in osteogenic lineage precursors. Increasing evidence supports that similar to normal development, the bone microenvironment (BME) underlies OS initiation and progression. Here, we recapitulate the physiological processes that regulate bone homeostasis and review the current knowledge about how OS cells and BME communicate and interact, describing how these interactions affect OS cell growth, metastasis, cancer stem cell fate and therapy outcome.


Asunto(s)
Huesos/fisiología , Microambiente Celular/fisiología , Homeostasis/fisiología , Modelos Biológicos , Metástasis de la Neoplasia/fisiopatología , Osteosarcoma/fisiopatología , Transducción de Señal , Humanos , Transducción de Señal/fisiología
14.
Stem Cells ; 32(9): 2529-38, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24806705

RESUMEN

As the nervous system exerts direct and indirect effects on stem cells mobilization and catecholamines mobilize hematopoietic stem cells, we hypothesized that dopamine might induce mesenchymal progenitor cells (MPCs) mobilization. We show that dopamine induced in vitro MPCs migration through D2-class receptors, and their alternative phosphoinositide 3-kinase/Akt pathways. Also, administration of catecholamines induced in vivo mobilization of colony-forming unit-fibroblast in mice. In contrast, in vitro and in vivo MPCs migration was suppressed by D2-class receptors antagonists and blocking antibodies, consistent with dopamine signaling pathway implication. In humans, patients treated with L-dopa or catecholaminergic agonists showed a significant increase of a MPC-like population (CD45-CD31-CD34-CD105+) in their peripheral blood. These findings reveal a new link between catecholamines and MPCs mobilization and suggest the potential use of D2-class receptors agonists for mobilization of MPCs in clinical settings.


Asunto(s)
Dopamina/farmacología , Células Madre Mesenquimatosas/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Dopamina/metabolismo , Femenino , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores Dopaminérgicos/metabolismo , Transducción de Señal/efectos de los fármacos
15.
Stem Cells ; 32(5): 1136-48, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24446210

RESUMEN

The cellular microenvironment plays a relevant role in cancer development. We have reported that mesenchymal stromal/stem cells (MSCs) deficient for p53 alone or together with RB (p53(-/-)RB(-/-)) originate leiomyosarcoma after subcutaneous (s.c.) inoculation. Here, we show that intrabone or periosteal inoculation of p53(-/-) or p53(-/-)RB(-/-) bone marrow- or adipose tissue-derived MSCs originated metastatic osteoblastic osteosarcoma (OS). To assess the contribution of bone environment factors to OS development, we analyzed the effect of the osteoinductive factor bone morphogenetic protein-2 (BMP-2) and calcified substrates on p53(-/-)RB(-/-) MSCs. We show that BMP-2 upregulates the expression of osteogenic markers in a WNT signaling-dependent manner. In addition, the s.c. coinfusion of p53(-/-)RB(-/-) MSCs together with BMP-2 resulted in appearance of tumoral osteoid areas. Likewise, when p53(-/-)RB(-/-) MSCs were inoculated embedded in a calcified ceramic scaffold composed of hydroxyapatite and tricalciumphosphate (HA/TCP), tumoral bone formation was observed in the surroundings of the HA/TCP scaffold. Moreover, the addition of BMP-2 to the ceramic/MSC implants further increased the tumoral osteoid matrix. Together, these data indicate that bone microenvironment signals are essential to drive OS development.


Asunto(s)
Neoplasias Óseas/patología , Huesos/patología , Microambiente Celular , Células Madre Mesenquimatosas/patología , Osteosarcoma/patología , Animales , Western Blotting , Proteína Morfogenética Ósea 2/farmacología , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Huesos/efectos de los fármacos , Huesos/metabolismo , Fosfatos de Calcio/química , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Cerámica/química , Durapatita/química , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Osteosarcoma/genética , Osteosarcoma/metabolismo , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Andamios del Tejido/química , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
16.
Mol Imaging ; 132014.
Artículo en Inglés | MEDLINE | ID: mdl-25248941

RESUMEN

Homing is an inherent, complex, multistep process performed by cells such as human bone marrow mesenchymal stem cells (hMSCs) to travel from a distant location to inflamed or damaged tissue and tumors. This ability of hMSCs has been exploited as a tumor-targeting strategy in cell-based cancer therapy. The purpose of this study was to investigate the applicability of 111In-oxine for tracking hMSCs in vivo by combining single-photon emission computed tomography (SPECT) and magnetic resonance imaging (MRI). 111In-labeled hMSCs (106 cells) were infused intraperitoneally in neuroblastoma-bearing mice, whereas a control group received a dose of free 111In-oxine. SPECT and MRI studies were performed 24 and 48 hours afterwards. Initially, the images showed similar activity in the abdomen in both controls and hMSC-injected animals. In general, abdominal activity decreases at 48 hours. hMSC-injected animals showed increased uptake in the tumor area at 48 hours, whereas the control group showed a low level of activity at 24 hours, which decreased at 48 hours. In conclusion, tracking 111In-labeled hMSCs combining SPECT and MRI is feasible and may be transferable to clinical research. The multimodal combination is essential to ensure appropriate interpretation of the images.


Asunto(s)
Células Madre Mesenquimatosas/diagnóstico por imagen , Neuroblastoma/diagnóstico por imagen , Compuestos Organometálicos/farmacocinética , Oxiquinolina/análogos & derivados , Animales , Línea Celular Tumoral , Células Cultivadas , Humanos , Imagen por Resonancia Magnética , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones SCID , Trasplante de Neoplasias , Neuroblastoma/patología , Oxiquinolina/farmacocinética , Tomografía Computarizada de Emisión de Fotón Único/métodos
17.
Cancer Res Commun ; 3(3): 347-360, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36875156

RESUMEN

Cancer immunotherapy aims to activate the immune system. Some immunotherapeutic agents can be loaded in carrier cells for delivering to the tumors. However, a challenge with cell-based therapies is the selection of the appropriate cells to produce effective clinical outcomes. We hypothesize that therapies based on cells presenting a natural low proinflammatory profile ("silent cells") in the peripheral blood would result in better antitumor responses by increasing their homing to the tumor site. We studied our hypothesis in an immunotherapy model consisting of mesenchymal stromal cells (MSCs) carrying oncolytic adenoviruses for the treatment of immunocompetent mice. Toll-like receptor signaling-deficient cells (TLR4, TLR9, or MyD88 knockout) were used as "silent cells," while regular MSCs were used as control. Although in vitro migration was similar in regular and knockout carrier cells, in vivo tumor homing of silent cells was significantly higher after systemic administration. This better homing to the tumor site was highly related to the mild immune response triggered by these silent cells in peripheral blood. As a result, the use of silent cells significantly improved the antitumor efficacy of the treatment in comparison with the use of regular MSCs. While cancer immunotherapies generally aim to boost local immune responses in the tumor microenvironment, low systemic inflammation after systemic administration of the treatment may indeed enhance their tumor homing and improve the overall antitumor effect. These findings highlight the importance of selecting appropriate donor cells as therapeutic carriers in cell-based therapies for cancer treatment. Significance: Cells carrying drugs, virus, or other antitumor agents are commonly used for the treatment of cancer. This research shows that silent cells are excellent carriers for immunotherapies, improving tumor homing and enhancing the antitumor effect.


Asunto(s)
Antineoplásicos , Viroterapia Oncolítica , Animales , Ratones , Transducción de Señal , Antineoplásicos/farmacología , Inmunoterapia , Receptores Toll-Like
18.
Stem Cells ; 29(2): 179-92, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21732477

RESUMEN

Human sarcomas have been modeled in mice by expression of specific fusion genes in mesenchymal stem cells (MSCs). However, sarcoma models based on human MSCs are still missing. We attempted to develop a model of liposarcoma by expressing FUS (FUsed in Sarcoma; also termed TLS, Translocated in LipoSarcoma)-CHOP (C/EBP HOmologous Protein; also termed DDIT3, DNA Damage-Inducible Transcript 3), a hallmark mixoid liposarcoma-associated fusion oncogene, in wild-type and p53-deficient mouse and human adipose-derived mesenchymal stem/stromal cells (ASCs). FUS-CHOP induced liposarcoma-like tumors when expressed in p53(-/-) but not in wild-type (wt) mouse ASCs (mASCs). In the absence of FUS-CHOP, p53(-/-) mASCs forms leiomyosarcoma, indicating that the expression of FUS-CHOP redirects the tumor genesis/phenotype. FUS-CHOP expression in wt mASCs does not initiate sarcomagenesis, indicating that p53 deficiency is required to induce FUS-CHOP-mediated liposarcoma in fat-derived mASCs. In a human setting, p53-deficient human ASCs (hASCs) displayed a higher in vitro growth rate and a more extended lifespan than wt hASCs. However, FUS-CHOP expression did not induce further changes in culture homeostasis nor initiated liposarcoma in either wt or p53-depleted hASCs. These results indicate that FUS-CHOP expression in a p53-deficient background is sufficient to initiate liposarcoma in mouse but not in hASCs, suggesting the need of additional cooperating mutations in hASCs. A microarray gene expression profiling has shed light into the potential deregulated pathways in liposarcoma formation from p53-deficient mASCs expressing FUS-CHOP, which might also function as potential cooperating mutations in the transformation process from hASCs.


Asunto(s)
Adipocitos/metabolismo , Liposarcoma/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción CHOP/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Adipocitos/citología , Animales , Diferenciación Celular , Proliferación Celular , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Liposarcoma/genética , Liposarcoma/patología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína FUS de Unión a ARN/genética , Proteínas Recombinantes de Fusión/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción CHOP/genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
19.
Adv Exp Med Biol ; 741: 187-205, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22457111

RESUMEN

The recognition of the therapeutic potential of Multipotent Mesenchymal Stromal Cells (MSCs) is one of the most exciting recent advances in cell therapy. In just ten years, since the description of the multilineage potential of MSCs by Pittenger et al in 1999 until now, MSCs are being used in more than 150 clinical trials as therapeutic agents. The potential of these cells for cell-based therapies relies on several key properties: (1) their capacity to differentiate into several cell lineages; (2) their lack of immunogenicity and their immunomodulatory properties; (3) their ex vivo expansion potential; (4) their ability to secrete soluble factors which regulate crucial biological functions such as proliferation and differentiation over a broad spectrum of target cells; and (5) their ability to home to damaged tissues and tumor sites. Based on these properties MSCs are being exploited worldwide for a wide range of potential clinical applications including cell replacement strategies, treatment of graft-versus-host disease, autoimmune diseases and rejection after solid organ transplantation as well as their use as vehicles to deliver anti-cancer therapies. Importantly, the low inherent immunogenicity of MSCs means that they could be used not only for autologous but also for allogeneic cell therapies. In addition, increasing evidence has revealed a complex relationship between MSCs and cancer. Thus, solid evidence has placed MSCs transformed with specific mutations as the most likely cell of origin for certain sarcomas, and MSCs have been reported to both, inhibit or promote tumor growth depending on yet undefined conditions. Here we will thoroughly discuss the different potential clinical applications of MSC as well as the role of MSCs on sarcomagenesis and the control of tumor growth.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Mesenquimatosas/fisiología , Células Madre Multipotentes/fisiología , Neoplasias/terapia , Enfermedades Autoinmunes/terapia , Biomarcadores/metabolismo , Ensayos Clínicos como Asunto , Rechazo de Injerto/terapia , Enfermedad Injerto contra Huésped/terapia , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Neoplasias/patología , Neoplasias/fisiopatología , Regeneración/fisiología
20.
Front Cell Dev Biol ; 10: 835273, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35693929

RESUMEN

We have observed a drug-tolerant/persister state in a human glioblastoma (GBM) cell line after exposure to temozolomide, the standard-of-care chemotherapeutic agent for GBM. We used a multicolor lentiviral genetic barcode labeling to follow cell population evolution during temozolomide treatment. We observed no change in the distribution of the different colored populations of cells in persister or resistant cells suggesting that pre-existing minor subpopulations, which would be expected to be restricted to a single color, were not amplified/selected during the response to the drug. We have previously identified four genes (CHI3L1, FAT2, KLK5, and HB-EGF) that were over-expressed during the persister stage. Single-cell analysis of these four genes indicated that they were expressed in different individual cells ruling out the existence of a single persister-specific clone but suggesting rather a global answer. Even so, the transitory silencing of CHI3L1, FAT2, or KLK5 influenced the expression of the other three genes and the survival of U251 cells in absence of temozolomide. Since proteins encoded by the four genes are all localized in the extracellular matrix or interact within the extracellular compartment, we propose that cellular interactions and communications are important during the persister stage before the acquisition of chemo-resistance. Thus, persisters might be a new therapeutically relevant target in GBM.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA