Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Angew Chem Int Ed Engl ; 58(10): 3067-3072, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30537383

RESUMEN

The selective delivery of therapeutic and imaging agents to tumoral cells has been postulated as one of the most important challenges in the nanomedicine field. Meta-iodobenzilguanidine (MIBG) is widely used for the diagnosis of neuroblastoma (NB) due to its strong affinity for the norepinephrine transporter (NET), usually overexpressed on the membrane of malignant cells. Herein, a family of novel Y-shaped scaffolds has been synthesized, which have structural analogues of MIBG covalently attached at each end of the Y-structure. The cellular uptake capacity of these double-targeting ligands has been evaluated in vitro and in vivo, yielding one specific Y-shaped structure that is able to be engulfed by the malignant cells, and accumulates in the tumoral tissue, at significantly higher levels than the structure containing only one single targeting agent. This Y-shaped ligand can provide a powerful tool for the current treatment and diagnosis of this disease.


Asunto(s)
3-Yodobencilguanidina/análogos & derivados , Portadores de Fármacos/química , Neuroblastoma/diagnóstico por imagen , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/análisis , 3-Yodobencilguanidina/metabolismo , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Portadores de Fármacos/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Ratones Endogámicos NOD , Ratones SCID , Modelos Moleculares , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Imagen Óptica
3.
Cytotherapy ; 20(11): 1337-1344, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30327248

RESUMEN

BACKGROUND: Bronchopulmonary dysplasia (BPD) is the most prevalent sequelae of premature birth, for which therapeutic options are currently limited. Mesenchymal stromal cells (MSCs) are a potential therapy for prevention or reversal of BPD. SERIES OF CASES: We report on two infants with severe BPD in whom off-label treatment with repeated intravenous doses of allogeneic bone marrow-derived MSCs were administered. We analyzed the temporal profile of serum and tracheal cytokines and growth factors as well as safety, tolerability and clinical response. The administration of repeated intravenous doses of MSCs in two human babies with severe and advanced BPD was feasible and safe and was associated with a decrease of pro-inflammatory molecules and lung injury biomarkers. Both patients were at very advanced stages of BPD with very severe lung fibrosis and did not survive the disease. CONCLUSIONS: MSCs are a promising therapy for BPD, but they should be administered in early stages of the disease.


Asunto(s)
Displasia Broncopulmonar/terapia , Pulmón/patología , Trasplante de Células Madre Mesenquimatosas/métodos , Administración Intravenosa , Biomarcadores/sangre , Displasia Broncopulmonar/diagnóstico por imagen , Citocinas/sangre , Femenino , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas , Tráquea/metabolismo
4.
Stem Cells ; 33(9): 2825-37, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26037670

RESUMEN

The genetic programs that maintain hematopoiesis during steady state in physiologic conditions are different from those activated during stress. Here, we show that hematopoietic stem cells (HSCs) with deficiencies in components of the alternative NFκB pathway (the NFκB inducing kinase, NIK, and the downstream molecule NFκB2) had a defect in response to stressors such as supraphysiological doses of cytokines, chemotherapy, and hematopoietic transplantation. NIK-deficient mice had peripheral blood and bone marrow leukocyte numbers within normal ranges (except for the already reported defects in B-cell maturation); however, HSCs showed significantly slower expansion capacity in in vitro cultures compared to wild-type HSCs. This was due to a delayed cell cycle and increased apoptosis. In vivo experiments showed that NIK-deficient HSCs did not recover at the same pace as controls when challenged with myeloablative chemotherapy. Finally, NIK-deficient HSCs showed a significantly decreased competitive repopulation capacity in vivo. Using HSCs from mice deficient in one of two downstream targets of NIK, that is, either NFκB2 or c-Rel, only NFκB2 deficiency recapitulated the defects detected with NIK-deficient HSCs. Our results underscore the role of NIK and the alternative NFκB pathway for the recovery of normal levels of hematopoiesis after stress.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Estrés Fisiológico/fisiología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad p52 de NF-kappa B/fisiología , Quinasa de Factor Nuclear kappa B
5.
Blood Cells Mol Dis ; 55(3): 220-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26227851

RESUMEN

We studied whether chemokines may have a role in relapses in childhood acute lymphoblastic leukemia (ALL). We compared the levels of chemokine receptors in marrow samples from 82 children with ALL at diagnosis versus 15 at relapses, and quantified the levels of chemokines in central system fluid (CSF) samples. The functional role of specific chemokines was studied in vitro and in vivo. The expression of some chemokine receptors was upregulated upon leukemic relapse, both in B- and in T-ALL, and in cases of medullary and extramedullary involvement. CXCL10 induced chemotaxis in leukemic cell lines and in primary leukemic cells, depending upon the levels of CXCR3 expression. CXCL10 specifically diminished chemotherapy-induced apoptosis on ALL cells expressing CXCR3, partially inhibiting caspase activation and maintaining the levels of the antiapoptotic protein Bcl-2. Finally, immunodeficient mice engrafted with CXCR3-expressing human leukemic cells showed decreased infiltration of marrow, spleen, and CNS after receiving a CXCR3-antagonist molecule. CXCR3 signaling in ALL may have a dual function: chemotactic for the localisation of leukemic blasts in specific niches, and it may also confer resistance to chemotherapy, enhancing the chances for relapses.


Asunto(s)
Antineoplásicos/farmacología , Quimiocinas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Quimiocina/metabolismo , Animales , Antineoplásicos/uso terapéutico , Quimiotaxis de Leucocito , Niño , Resistencia a Antineoplásicos , Humanos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Recurrencia
6.
Lung ; 193(1): 19-23, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25355250

RESUMEN

PURPOSE: Traditional inflammatory biomarkers are insufficient for the evaluation of bronchiolitis severity. Recent investigations have shown that the receptor for advanced glycation end product (RAGE) and its soluble isoforms (sRAGE) play a critical role in the pathogenesis of lung injury. Main objective was to assess the serum levels of sRAGE of children with severe bronchiolitis admitted to the pediatric intensive care unit (PICU). Secondary objective was to study sRAGE correlation with the evolution and traditional biomarkers. METHODS: Prospective, observational and descriptive study, 43 healthy controls and 37 patients (December 2011-February 2012) were enrolled. sRAGE levels were assessed and compared. In patients, the relation between sRAGE levels and clinical evolution, respiratory assistance, white blood cell count, absolute neutrophils count, serum C-reactive protein, and serum procalcitonin was analyzed. RESULTS: A statistical difference was found in the mean value of sRAGE at PICU admission between patients and controls (1,215.7 ± 535 vs 849 ± 579 pg/ml). Also a significant inverse correlation was found between sRAGE and the Wood-Downes Score at admission (p = 0.02). CONCLUSIONS: Serum sRAGE could be elevated in children with bronchiolitis. Larger clinical studies are necessary to elucidate its role as a bronchiolitis inflammatory and/or lung injury biomarker.


Asunto(s)
Bronquiolitis/sangre , Receptores Inmunológicos/sangre , Factores de Edad , Biomarcadores/sangre , Bronquiolitis/diagnóstico , Estudios de Casos y Controles , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Unidades de Cuidado Intensivo Pediátrico , Tiempo de Internación , Masculino , Admisión del Paciente , Proyectos Piloto , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Receptor para Productos Finales de Glicación Avanzada , Índice de Severidad de la Enfermedad , Factores de Tiempo , Regulación hacia Arriba
7.
Brain Inj ; 29(12): 1497-510, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26287760

RESUMEN

OBJECTIVE: Traumatic brain injury (TBI) is the leading cause of mortality and morbidity in paediatric patients after the first year of life. The aim of this study was to evaluate effects of locally administered allogeneic mesenchymal stem cells (MSC), in the acute period after a TBI. METHODOLOGY: MSC were isolated from peritoneal fat of healthy rats, expanded in vitro and labelled with the green fluorescent protein. Rats were placed in one of three experimental groups: (1) CONTROL: TBI, (2) IP-CONTROL: TBI + local saline and (3) IP-Treat: TBI + 2 × 10(5) MSC 24 hours after receiving a moderate, unilateral, controlled cortical impact. Motor and cognitive behavioural tests were performed to evaluate functional recovery. Histological examination and immunohistochemistry were used to identify cell distribution. MAIN RESULTS: Improved performance was found on motor tests in the MSC-treated group compared to control groups. MSC were found in the perilesional area and their number decreased with time after transplantation. MSC treatment increased the cell density in the hippocampus (CA3 pyramidal cells and granule cells in the dentate gyrus) and enhanced neurogenesis in this area. CONCLUSION: MSC cell therapy resulted in better recovery of motor function compared with the control group. This cellular therapy might be considered for patients suffering from TBI.


Asunto(s)
Tejido Adiposo/trasplante , Lesiones Encefálicas/terapia , Animales , Células de la Médula Ósea , Encéfalo/patología , Giro Dentado/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Modelos Animales , Neurogénesis/fisiología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/fisiología
8.
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
9.
Mol Ther ; 18(12): 2173-81, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20736931

RESUMEN

Lineage-specific differentiation potential varies among different human pluripotent stem cell (hPSC) lines, becoming therefore highly desirable to prospectively know which hPSC lines exhibit the highest differentiation potential for a certain lineage. We have compared the hematopoietic potential of 14 human embryonic stem cell (hESC)/induced pluripotent stem cell (iPSC) lines. The emergence of hemogenic progenitors, primitive and mature blood cells, and colony-forming unit (CFU) potential was analyzed at different time points. Significant differences in the propensity to differentiate toward blood were observed among hPSCs: some hPSCs exhibited good blood differentiation potential, whereas others barely displayed blood-differentiation capacity. Correlation studies revealed that the CFU potential robustly correlates with hemogenic progenitors and primitive but not mature blood cells. Developmental progression of mesoendodermal and hematopoietic transcription factors expression revealed no correlation with either hematopoietic initiation or maturation efficiency. Microarray studies showed distinct gene expression profile between hPSCs with good versus poor hematopoietic potential. Although neuroectoderm-associated genes were downregulated in hPSCs prone to hematopoietic differentiation many members of the Nodal/Activin signaling were upregulated, suggesting that this signaling predicts those hPSC lines with good blood-differentiation potential. The association between Nodal/Activin signaling and the hematopoietic differentiation potential was confirmed using loss- and gain-of-function functional assays. Our data reinforce the value of prospective comparative studies aimed at determining the lineage-specific differentiation potential among different hPSCs and indicate that Nodal/Activin signaling seems to predict those hPSC lines prone to hematopoietic specification.


Asunto(s)
Activinas/metabolismo , Diferenciación Celular , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes/citología , Transducción de Señal , Activinas/farmacología , Linaje de la Célula , Perfilación de la Expresión Génica , Humanos
10.
Regen Med ; 15(9): 2053-2065, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-33245008

RESUMEN

Venous leg ulcers (VLU) represent an uphill economic, health and social burden, aggravated in the elderly. Best-practice care interventions are often insufficient and alternative therapies need to be explored. Herein, we have treated for the first time a chronic VLU in an elderly patient by combining cell therapy and tissue engineering in the context of a compassionate use. The administration of allogeneic adipose-derived mesenchymal stromal cells (MSCs) embedded in a plasma-based bioengineered dermis covering the ulcer bed and also injected into the ulcer margins led to the complete closure of a 10-year recalcitrant VLU in an 85-year-old patient. Regenerative properties of MSCs might be boosted by the use of bioengineered matrices for their delivery.


Asunto(s)
Úlcera de la Pierna , Células Madre Mesenquimatosas , Úlcera Varicosa , Tejido Adiposo , Anciano , Anciano de 80 o más Años , Humanos , Úlcera de la Pierna/terapia , Ingeniería de Tejidos
11.
Cancers (Basel) ; 12(5)2020 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-32354143

RESUMEN

Little is known about the effect of oncolytic adenovirotherapy on pediatric tumors. Here we present the clinical case of a refractory neuroblastoma that responded positively to Celyvir (ICOVIR-5 oncolytic adenovirus delivered by autologous mesenchymal stem cells) for several months. We analyzed samples during tumor evolution in order to identify molecular and mutational features that could explain the interactions between treatment and tumor and how the balance between both of them evolved. We identified a higher adaptive immune infiltration during stabilized disease compared to progression, and also a higher mutational rate and T-cell receptor (TCR) diversity during disease progression. Our results indicate an initial active role of the immune system controlling tumor growth during Celyvir therapy. The tumor eventually escaped from the control exerted by virotherapy through acquisition of resistance by the tumor microenvironment that exhausted the initial T cell response.

12.
Pediatr Pulmonol ; 55(12): 3429-3436, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32852101

RESUMEN

INTRODUCTION AND OBJECTIVES: Acute bronchiolitis (AB) is the leading cause of hospitalization in infants and around 5% require intensive care treatment. Early identification of children diagnosed with AB at a high risk of severe progression is of great interest. The receptor for advanced glycation end products (RAGE), highly expressed in lung tissue, regulates immune responses and inflammation, and its soluble form, sRAGE, is believed to have an anti-inflammatory role. We hypothesized serum sRAGE might be a major determinant of AB severity and prognosis. This study was conducted to measure serum sRAGE in infants with severe AB and to assess its correlation with clinical severity, immediate complications, and outcome. METHODS: Single-center, prospective, observational study of hospitalized children with severe bronchiolitis admitted to the Pediatric Intensive Care Unit (PICU), from September 2015 to September 2016. RESULTS: A total of 52 children and 27 controls were included. The cases age ranged from 11 days to 21 months, resulting in a significant age difference with controls (11.85 vs 4.84 months, P < .01). Serum levels of sRAGE were lower but not significant in severe AB patients than in controls (1350.93 vs 1450.42 pg/mL; P = .399). No correlation was found between serum sRAGE and causative viruses, clinical symptoms, Wood-Downes score (a clinical severity score) on admission, respiratory support, or length of hospital stay. Serum sRAGE was also lower in the cases having had a previous respiratory disease (1463.84 vs 1072.43 pg/mL; P = .049). However, it was higher in patients with any lung consolidation on the chest X-ray (1584.79 vs 1131.62 pg/mL; P = .044) and weakly positively correlated with classical biomarkers (maximum C-reactive protein, +0.295, P = .034; maximum procalcitonin, +0.309; P = .029). CONCLUSION: This single-center study reveals that sRAGE couldn't predict AB severity or outcome in children hospitalized at PICU. Nevertheless, it significantly increased in the presence of any lung consolidation and had a positive correlation with classical biomarkers. The utility of sRAGE in this population could be probably elucidated with a better understanding of AGE-RAGE axis.


Asunto(s)
Bronquiolitis/diagnóstico , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Biomarcadores/metabolismo , Bronquiolitis/metabolismo , Proteína C-Reactiva , Niño , Femenino , Hospitalización , Humanos , Lactante , Inflamación , Unidades de Cuidado Intensivo Pediátrico , Tiempo de Internación , Pulmón/metabolismo , Masculino , Pronóstico , Estudios Prospectivos
13.
Front Med (Lausanne) ; 7: 576558, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324660

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is an incurable inherited mucocutaneous fragility disorder characterized by recurrent blisters, erosions, and wounds. Continuous blistering triggers overlapping cycles of never-ending healing and scarring commonly evolving to chronic systemic inflammation and fibrosis. The systemic treatment with allogeneic mesenchymal cells (MSC) from bone marrow has previously shown benefits in RDEB. MSC from adipose tissue (ADMSC) are easier to isolate. This is the first report on the use of systemic allogeneic ADMSC, correlating the clinical, inflammatory, and immunologic outcomes in RDEB indicating long-lasting benefits. We present the case of an RDEB patient harboring heterozygous biallelic COL7A1 gene mutations and with a diminished expression of C7. The patient presented with long-lasting refractory and painful oral ulcers distressing her quality of life. Histamine receptor antagonists, opioid analgesics, proton-pump inhibitors, and low-dose tricyclic antidepressants barely improved gastric symptoms, pain, and pruritus. Concomitantly, allogeneic ADMSC were provided as three separate intravenous injections of 106 cells/kg every 21 days. ADMSC treatment was well-tolerated. Improvements in wound healing, itch, pain and quality of life were observed, maximally at 6-9 months post-treatment, with the relief of symptoms still noticeable for up to 2 years. Remarkably, significant modifications in PBL participating in both the innate and adaptive responses, alongside regulation of levels of profibrotic factors, MCP-1/CCL2 and TGF-ß, correlated with the health improvement. This treatment might represent an alternative for non-responding patients to conventional management. It seems critical to elucidate the paracrine modulation of the immune system by MSC for their rational use in regenerative/immunoregulatory therapies.

14.
Carcinogenesis ; 30(9): 1628-37, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19587093

RESUMEN

MLL rearrangements are hallmark genetic abnormalities in infant leukemia known to arise in utero. They can be induced during human prenatal development upon exposure to etoposide. We also hypothesize that chronic exposure to etoposide might render cells more susceptible to other genomic insults. Here, for the first time, human embryonic stem cells (hESCs) were used as a model to test the effects of etoposide on human early embryonic development. We addressed whether: (i) low doses of etoposide promote MLL rearrangements in hESCs and hESCs-derived hematopoietic cells; (ii) MLL rearrangements are sufficient to confer hESCs with a selective growth advantage and (iii) continuous exposure to low doses of etoposide induces hESCs to acquire other chromosomal abnormalities. In contrast to cord blood-derived CD34(+) and hESC-derived hematopoietic cells, exposure of undifferentiated hESCs to a single low dose of etoposide induced a pronounced cell death. Etoposide induced MLL rearrangements in hESCs and their hematopoietic derivatives. After long-term culture, the proportion of hESCs harboring MLL rearrangements diminished and neither cell cycle variations nor genomic abnormalities were observed in the etoposide-treated hESCs, suggesting that MLL rearrangements are insufficient to confer hESCs with a selective proliferation/survival advantage. However, continuous exposure to etoposide induced MLL breaks and primed hESCs to acquire other major karyotypic abnormalities. These data show that chronic exposure of developmentally early stem cells to etoposide induces MLL rearrangements and make hESCs more prone to acquire other chromosomal abnormalities than postnatal CD34(+) cells, linking embryonic genotoxic exposure to genomic instability.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Aberraciones Cromosómicas/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Etopósido/toxicidad , Reordenamiento Génico , Proteína de la Leucemia Mieloide-Linfoide/genética , Antígenos CD34/análisis , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Madre Embrionarias/ultraestructura , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina , Humanos
15.
J Pediatr Endocrinol Metab ; 31(6): 649-654, 2018 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-29723156

RESUMEN

BACKGROUND: The receptor for advanced glycation end products (RAGE) has a critical role in the pathogenesis of inflammation. In healthy children, its basal expression on the peripheral blood mononuclear cell (PBMC) and the basal circulating soluble RAGE (sRAGE) levels are unknown. The aim of this study was to describe both. METHODS: This is a monocentric, observational and descriptive study of samples obtained from healthy children. The RAGE expression on PBMC was analyzed using flow cytometry. The sRAGE values were determined with a specific sandwich enzyme-linked immunosorbent assay (ELISA) kit, later the relation between cellular RAGE and sRAGE was described. RESULTS: Forty-three children were included. The median sRAGE level was 849.0±579.0 pg/mL. The RAGE mean fluorescence intensity (MFI) was 1382±506 in monocytes and 792±506 in lymphocytes. There were no differences between genders. A negative correlation was found between sRAGE and RAGE MFI in lymphocytes (r=-0.3; p=0.04). CONCLUSIONS: We describe for the first time the RAGE surface levels on PBMC in children. It showed a negative correlation with sRAGE. The sRAGE circulating level is lower than the sRAGE level described in adult population or non-healthy children. Our findings should be confirmed in order to apply them as reference values for future investigations.


Asunto(s)
Leucocitos Mononucleares/metabolismo , Receptor para Productos Finales de Glicación Avanzada/sangre , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Adolescente , Factores de Edad , Envejecimiento/sangre , Envejecimiento/metabolismo , Antígenos de Superficie/metabolismo , Niño , Preescolar , Femenino , Citometría de Flujo , Voluntarios Sanos , Humanos , Masculino , Isoformas de Proteínas/sangre , Isoformas de Proteínas/metabolismo , Solubilidad
16.
J Leukoc Biol ; 103(5): 965-971, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29485692

RESUMEN

The CD64 receptor has been described as a biomarker of bacterial infection. We speculated that CD64 surface expression on monocytes and granulocytes of children with severe acute bronchiolitis (SAB) could be altered in cases of probable bacterial infection (PBI) determined using classical biomarkers (procalcitonin and C-reactive protein, leukocyte count, and radiographic findings). A prospective observational pilot study was conducted from October 2015 to February 2016 in children admitted for pediatric critical care. A blood sample was taken in the first 24 hours of admission, and CD64 was measured by flow cytometry. The values obtained were analyzed and correlated with traditional biomarkers of PBI. Thirty-two children were included; a correlation was found between CD64 expression and the PBI criteria. CD64 surface expression was higher in children with PBI (area under the receiver operating characteristic curve of 0.73; P = 0.042) and the percentage of CD64+ granulocytes was higher in children with PBI. This is the first study to describe CD64 surface expression on monocytes and granulocytes in SAB, finding CD64 values to be higher in children with PBI. Larger clinical studies are needed to elucidate the real accuracy of CD64 as a biomarker of bacterial infection.


Asunto(s)
Infecciones Bacterianas/diagnóstico , Biomarcadores/metabolismo , Bronquiolitis/complicaciones , Granulocitos/metabolismo , Monocitos/metabolismo , Receptores de IgG/metabolismo , Enfermedad Aguda , Infecciones Bacterianas/etiología , Infecciones Bacterianas/metabolismo , Bronquiolitis/microbiología , Niño , Femenino , Granulocitos/inmunología , Humanos , Masculino , Monocitos/inmunología , Proyectos Piloto , Estudios Prospectivos , Índice de Severidad de la Enfermedad
17.
Mol Syst Biol ; 1: 2005.0028, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16729063

RESUMEN

Translating a graded morphogen distribution into tight response borders is central to all developmental processes. Yet, the molecular mechanisms generating such behavior are poorly understood. During patterning of the Drosophila embryonic ventral ectoderm, a graded mitogen-activated protein kinase (MAPK) activation is converted into an all-or-none degradation switch of the Yan transcriptional repressor. Replacing the cardinal phosphorylated amino acid of Yan by a phosphomimetic residue allowed its degradation in a MAPK-independent manner, consistent with Yan phosphorylation being the critical event in generating the switch. Several alternative threshold mechanisms that could, in principle, be realized by this phosphorylation, including first order, cooperativity, positive feedback and zero-order ultrasensitivity, were analyzed. We found that they can be distinguished by their kinetics and steady-state responses to Yan overexpression. In agreement with the predictions for zero-order kinetics, an increase in Yan levels did not shift the degradation border, but significantly elevated the time required to reach steady state. We propose that a reversible loop of Yan phosphorylation implements a zero-order ultrasensitivity-like threshold mechanism, with the capacity to form sharp thresholds that are independent of the level of Yan.


Asunto(s)
Tipificación del Cuerpo/fisiología , Biología Computacional , Proteínas de Drosophila/fisiología , Drosophila melanogaster/embriología , Proteínas del Ojo/fisiología , Regulación del Desarrollo de la Expresión Génica , Modelos Biológicos , Procesamiento Proteico-Postraduccional/fisiología , Proteínas Represoras/fisiología , Sustitución de Aminoácidos , Animales , Tipificación del Cuerpo/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ectodermo/fisiología , Ectodermo/ultraestructura , Embrión no Mamífero/fisiología , Embrión no Mamífero/ultraestructura , Activación Enzimática , Factor de Crecimiento Epidérmico/fisiología , Receptores ErbB/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Cinética , Sistema de Señalización de MAP Quinasas , Matemática , Proteínas de la Membrana/fisiología , Morfogénesis/genética , Morfogénesis/fisiología , Péptido Hidrolasas/metabolismo , Fosforilación
18.
Cancer Lett ; 371(2): 161-70, 2016 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-26655276

RESUMEN

We report here our clinical experience of a program of compassionate use of Celyvir--autologous marrow-derived mesenchymal stem cells (MSCs) carrying an oncolytic adenovirus--for treating children with advanced metastatic neuroblastoma. Children received weekly doses of Celyvir with no concomitant treatments. The tolerance was excellent, with very mild and self-limited viral-related symptoms. Patients could be distinguished based on their response to therapy: those who had a clinical response (either complete, partial or stabilization) and those who did not respond. We found differences between patients who responded versus those who did not when analyzing their respective MSCs, at the expression levels of adhesion molecules (CCR1, CXCR1 and CXCR4) and in migration capacities in transwell assays, and in immune-related molecules (IFNγ, HLA-DR). These results suggest interpatient differences in the homing and immune modulation capacities of the therapy administered. In addition, the pretherapy immune T cell status and the T effector response were markedly different between responders and non-responders. We conclude that multidoses of Celyvir have an excellent safety profile in children with metastatic neuroblastoma. Intrinsic patients' and MSCs' factors appear to be related to clinical outcome.


Asunto(s)
Adenoviridae/patogenicidad , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/virología , Neuroblastoma/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/patogenicidad , Biomarcadores/metabolismo , Adhesión Celular , Línea Celular , Movimiento Celular , Ensayos de Uso Compasivo , Femenino , Antígenos HLA-DR/metabolismo , Humanos , Interferón gamma/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Neuroblastoma/secundario , Neuroblastoma/virología , Viroterapia Oncolítica/efectos adversos , Fenotipo , Receptores CCR1/metabolismo , Receptores CXCR4/metabolismo , Receptores de Interleucina-8A/metabolismo , España , Factores de Tiempo , Trasplante Autólogo , Resultado del Tratamiento , Replicación Viral
19.
J Mater Chem B ; 3(24): 4831-4842, 2015 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-32262672

RESUMEN

Novel targeting agents against neuroblastoma based on the meta-iodobenzylguanidine (MIBG) moiety were synthesized and biologically evaluated for nanocarrier vectorization. These compounds have been anchored on the surface of drug loaded mesoporous silica nanocarriers, resulting in the improved cellular uptake in tumoral cells. Neuroblastoma (NB) is the most frequent extracranial pediatric tumor. Advanced forms of the disease (metastatic and/or refractory) have a dismal prognosis despite the combination of chemotherapy, radiotherapy, surgery and bone narrow transplants. These treatments carry severe side effects and, in some cases, compromise the life of the patient. MIBG has been widely applied in the medical diagnosis of NB due to its affinity for tumor cells through the norepinephrine transporter (NET), which is expressed in 90% of NB tumors. The exclusive accumulation of MIBG in neuroblastoma has been widely studied; however, its properties have been never exploited as a targeting agent in nanocarrier drug delivery systems. Several structural analogues of MIBG have been prepared and attached on the surface of nanocarriers. Their selective internalization has been tested against human neuroblastoma cells, which show, in the best case, cellular uptake four times higher than that of the naked nanosystem. Furthermore, in vivo experiments showed preferential and selective accumulation and retention of the targeted nanosystem comparing with the naked and only PEGylated counterpart systems. This novel nanosystem could be easily applicable to all kinds of drug delivery nanocarriers, providing a universal tool for neuroblastoma chemotherapies that is superior to classical approaches through a novel nanosystem exclusively designed to target this terrible malignancy.

20.
Oncolytic Virother ; 4: 149-55, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27512678

RESUMEN

Oncolytic virotherapy is gaining interest in the clinic as a new weapon against cancer. In vivo administration of oncolytic viruses showed important limitations that decrease their effectiveness very significantly: the antiviral immune response causes the elimination of the therapeutic effect, and the poor natural ability of oncolytic viruses to infect micrometastatic lesions significantly minimizes the effective dose of virus. This review will focus on updating the technical and scientific foundations of one of the strategies developed to overcome these limitations, ie, using cells as vehicles for oncolytic viruses. Among many candidates, a special type of adult stem cell, mesenchymal stem cells (MSCs), have already been used in the clinic as cell vehicles for oncolytic viruses, partly due to the fact that these cells are actively being evaluated for other indications. MSC carrier cells are used as Trojan horses loaded with oncoviruses, are administered systemically, and release their cargos at the right places. MSCs are equipped with an array of molecules involved in cell arrest in the capillaries (integrins and selectins), migration toward specific parenchymal locations within tissues (chemokine receptors), and invasion and degradation of the extracellular matrix (proteases). In addition to anatomical targeting capacity, MSCs have a well-recognized role in modulating immune responses by affecting cells of the innate (antigen-presenting cells, natural killer cells) and adaptive immune system (effector and regulatory lymphocytes). Therefore, carrier MSCs may also modulate the immune responses taking place after therapy, ie, the antiviral and the antitumor immune responses.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA