Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Genet ; 16(5): e1008815, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32453722

RESUMEN

Trimethylguanosine synthase 1 (TGS1) is a conserved enzyme that mediates formation of the trimethylguanosine cap on several RNAs, including snRNAs and telomerase RNA. Previous studies have shown that TGS1 binds the Survival Motor Neuron (SMN) protein, whose deficiency causes spinal muscular atrophy (SMA). Here, we analyzed the roles of the Drosophila orthologs of the human TGS1 and SMN genes. We show that the Drosophila TGS1 protein (dTgs1) physically interacts with all subunits of the Drosophila Smn complex (Smn, Gem2, Gem3, Gem4 and Gem5), and that a human TGS1 transgene rescues the mutant phenotype caused by dTgs1 loss. We demonstrate that both dTgs1 and Smn are required for viability of retinal progenitor cells and that downregulation of these genes leads to a reduced eye size. Importantly, overexpression of dTgs1 partially rescues the eye defects caused by Smn depletion, and vice versa. These results suggest that the Drosophila eye model can be exploited for screens aimed at the identification of genes and drugs that modify the phenotypes elicited by Tgs1 and Smn deficiency. These modifiers could help to understand the molecular mechanisms underlying SMA pathogenesis and devise new therapies for this genetic disease.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/crecimiento & desarrollo , Proteínas de Unión al ARN/genética , Proteínas del Complejo SMN/genética , Animales , Regulación hacia Abajo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Letales , Tamaño de los Órganos , Proteínas de Unión al ARN/metabolismo , Proteínas del Complejo SMN/metabolismo
2.
iScience ; 27(6): 109960, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38832015

RESUMEN

Limited information exists regarding the impact of interferons (IFNs) on the information carried by extracellular vesicles (EVs). This study aimed at investigating whether IFN-α2b, IFN-ß, IFN-γ, and IFN-λ1/2 modulate the content of EVs released by primary monocyte-derived macrophages (MDM). Small-EVs (sEVs) were purified by size exclusion chromatography from supernatants of MDM treated with IFNs. To characterize the concentration and dimensions of vesicles, nanoparticle tracking analysis was used. SEVs surface markers were examined by flow cytometry. IFN treatments induced a significant down-regulation of the exosomal markers CD9, CD63, and CD81 on sEVs, and a significant modulation of some adhesion molecules, major histocompatibility complexes and pro-coagulant proteins, suggesting IFNs influence biogenesis and shape the immunological asset of sEVs. SEVs released by IFN-stimulated MDM also impact lymphocyte function, showing significant modulation of lymphocyte activation and IL-17 release. Altogether, our results show that sEVs composition and activity are affected by IFN treatment of MDM.

3.
Blood ; 118(2): 380-9, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-21602525

RESUMEN

Although many literature data are available on the role of Notch signaling in T-cell acute lymphoblastic leukemia (ALL) biology, the importance of this molecular pathway in the development of B-lineage ALL (B-ALL) cells in the BM microenvironment is unknown so far. In this study, we used anti-Notch molecules neutralizing Abs and γ-secretase inhibitor (GSI) XII to investigate the role of the Notch signaling pathway in the promotion of human B-ALL cell survival in presence of stromal cell support. The treatment with combinations of anti-Notch molecule neutralizing Abs resulted in the decrease of B-ALL cell survival, either cultured alone or cocultured in presence of stromal cells from normal donors and B-ALL patients. Interestingly, the inhibition of Notch-3 and -4 or Jagged-1/-2 and DLL-1 resulted in a dramatic increase of apoptotic B-ALL cells by 3 days, similar to what is obtained by blocking all Notch signaling with the GSI XII. Our data suggest that the stromal cell-mediated antiapoptotic effect on B- ALL cells is mediated by Notch-3 and -4 or Jagged-1/-2 and DLL-1 in a synergistic manner.


Asunto(s)
Apoptosis/genética , Células de la Médula Ósea/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Proto-Oncogénicas/fisiología , Receptores Notch/fisiología , Células del Estroma/fisiología , Linfocitos B/patología , Células de la Médula Ósea/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/fisiología , Comunicación Celular/genética , Comunicación Celular/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteína Jagged-1 , Proteína Jagged-2 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptor Notch3 , Receptor Notch4 , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal/genética , Transducción de Señal/fisiología , Células del Estroma/metabolismo , Células Tumorales Cultivadas
4.
Free Radic Biol Med ; 208: 718-727, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37739138

RESUMEN

Physical Exercise (EXR) has been shown to have numerous beneficial effects on various systems in the human body. It leads to a decrease in the risk of mortality from chronic diseases, such as cardiovascular disease, cancer, metabolic and central nervous system disorders. EXR results in improving cardiovascular fitness, cognitive function, immune activity, endocrine action, and musculoskeletal health. These positive effects make EXR a valuable intervention for promoting overall health and well-being in individuals of all ages. These beneficial effects are partially mediated by the role of the regular EXR in the adaptation to redox homeostasis counteracting the sudden increase of ROS, the hallmark of many chronic diseases. EXR can trigger the release of numerous humoral factors, e.g. protein, microRNA (miRs), and DNA, that can be shuttled as cargo of Extracellular vesicles (EVs). EVs show different cargo modification after oxidative stress stimuli as well as after EXR. In this review, we aim to highlight the main studies on the role of EVs released during EXR and oxidative stress conditions in enhancing the antioxidant enzymes pathway and in the decrease of oxidative stress environment mediated by their cargo.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Humanos , Estrés Oxidativo , Vesículas Extracelulares/metabolismo , Ejercicio Físico , MicroARNs/genética , MicroARNs/metabolismo , Enfermedad Crónica
5.
Free Radic Biol Med ; 204: 266-275, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37182793

RESUMEN

Considering the role of redox homeostasis in exercise-induced signaling and adaptation, this study focuses on the exercise training-related intercellular communication of redox status mediated by circulating extracellular vesicles (EVs). 19 healthy young males were divided into trained (TG, 7) and untrained (UG, 12) subjects based on their VO2MAX. The UG subjects were further randomly distributed in experimental (UGEX, N = 7) and control (UGCTRL, N = 5) groups. The steady state of plasma EVs in TG and UGEX have been characterized for total number and size, as well as cargo redox status (antioxidants, transcription factors, HSPs) before, 3 and 24 h after a single bout of aerobic exercise (30', 70% HRM). Plasma EVs from UGEX and UGCTRL have been further characterized after 24 h from the last session of a 5-day consecutive aerobic training or no training, respectively. No differences were detected in the EVs' size and distribution at baseline in TG and UGEX (p>0.05), while the EVs cargo of UGEX showed a significantly higher concentration of protein carbonyl, Catalase, SOD2, and HSF1 compared to TG (p<0.05). 5 days of consecutive aerobic training in UGEX did not determine major changes in the steady-state number and size of EVs. The post-training levels of protein carbonyl, HSF1, Catalase, and SOD2 in EVs cargo of UGEX resulted significantly lower compared with UGEX before training and UGCTRL, resembling the steady-state levels in circulating EVs of TG subjects. Altogether, these preliminary data indicate that individual aerobic capacity influences the redox status of circulating EVs, and that short-term aerobic training impacts the steady-state redox status of EVs. Taking this pilot study as a paradigm for physio-pathological stimuli impacting redox homeostasis, our results offer new insights into the utilization of circulating EVs as biomarkers of exercise efficacy and of early impairment of oxidative-stress related diseases.


Asunto(s)
Ejercicio Físico , Vesículas Extracelulares , Masculino , Humanos , Catalasa/metabolismo , Proyectos Piloto , Oxidación-Reducción , Vesículas Extracelulares/metabolismo
6.
Redox Biol ; 63: 102737, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37236143

RESUMEN

Cardiovascular diseases (CVD) can cause various conditions, including an increase in reactive oxygen species (ROS) levels that can decrease nitric oxide (NO) availability and promote vasoconstriction, leading to arterial hypertension. Physical exercise (PE) has been found to be protective against CVD by helping to maintain redox homeostasis through a decrease in ROS levels, achieved by increased expression of antioxidant enzymes (AOEs) and modulation of heat shock proteins (HSPs). Extracellular vesicles (EVs) circulating in the body are a major source of regulatory signals, including proteins and nucleic acids. Interestingly, the cardioprotective role of EVs released after PE has not been fully described. The aim of this study was to investigate the role of circulating EVs, obtained through Size Exclusion Chromatography (SEC) of plasma samples from healthy young males (age: 26.95 ± 3.07; estimated maximum oxygen consumption rate (VO2max): 51.22 ± 4.85 (mL/kg/min)) at basal level (Pre_EVs) and immediately after a single bout of endurance exercise (30' treadmill, 70% heart rate (HR) -Post_EVs). Gene ontology (GO) analysis of proteomic data from isolated EVs, revealed enrichment in proteins endowed with catalytic activity in Post_EVs, compare to Pre_EVs, with MAP2K1 being the most significantly upregulated protein. Enzymatic assays on EVs derived from Pre and Post samples showed increment in Glutathione Reductase (GR) and Catalase (CAT) activity in Post_EVs. At functional level, Post_EVs, but not Pre_EVs, enhanced the activity of antioxidant enzymes (AOEs) and reduced oxidative damage accumulation in treated human iPS-derived cardiomyocytes (hCM) at basal level and under stress conditions (Hydrogen Peroxide (H2O2) treatment), resulting in a global cardioprotective effect. In conclusion, our data demonstrated, for the first time, that a single 30-min endurance exercise is able to alter the cargo of circulating EVs, resulting in cardioprotective effect through antioxidant activity.


Asunto(s)
Enfermedades Cardiovasculares , Vesículas Extracelulares , Masculino , Humanos , Adulto Joven , Adulto , Antioxidantes/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Peróxido de Hidrógeno/metabolismo , Proteómica , Enfermedades Cardiovasculares/metabolismo
7.
Stem Cells ; 29(6): 1001-11, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21563279

RESUMEN

Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are stromal precursors endowed with extensive immunomodulative properties. In this study, we aimed to assess whether Toll-like receptor-3 (TLR3)- and TLR4-activated BM-MSC influence human neutrophil (PMN) responses under coculture conditions. We show that TLR3 triggering by polyinosinic:polycytidylic acid dramatically amplifies, in a more significant manner than TLR4 triggering by lipopolysaccharide, the antiapoptotic effects that resting BM-MSC constitutively exert on PMN under coculture conditions, preserving a significant fraction of viable and functional PMN up to 72 hours. In addition, TLR3- and TLR4-activated BM-MSC enhance respiratory burst ability and CD11b expression by PMN. The coculture in the absence of cell contact and the incubation of PMN in supernatants harvested from TLR3- and TLR4-activated BM-MSC yield comparable results in terms of increased survival and immunophenotypic changes, thus suggesting the involvement of endogenous soluble factors. Neutralizing experiments reveal that the biological effects exerted on PMN by TLR3-activated BM-MSC are mediated by the combined action of interleukin 6, interferon-ß (IFN-ß), and granulocyte macrophage colony-stimulating factor (GM-CSF), while those exerted by TLR4-activated BM-MSC mostly depend on GM-CSF. MSC isolated from thymus, spleen, and subcutaneous adipose tissue behaves similarly. Finally, the effects exerted by TLR3- or TLR4-stimulated BM-MSC on PMN are conserved even after the previous priming of BM-MSC with IFN-γ and tumor necrosis factor-α. Our data highlight a novel mechanism by which MSC sustain and amplify the functions of PMN in response to TLR3- and TLR4-triggering and may consequently contribute to inflammatory disorders.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Neutrófilos/fisiología , Receptor Toll-Like 3/metabolismo , Células de la Médula Ósea/citología , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Interferón beta/metabolismo , Interferón gamma/farmacología , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Fenotipo , Poli I-C/farmacología , Receptor Toll-Like 3/agonistas , Factor de Necrosis Tumoral alfa/farmacología
8.
Oxid Med Cell Longev ; 2021: 1938492, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34853628

RESUMEN

Regular physical activity can enhance immune function and effectively prevents the spread of the cytokine response, thus reducing systemic low-grade inflammation and improving various immune markers. Moreover, regular exercise maintains redox homeostasis in skeletal muscle and other tissues, including immune cells, but the interconnection between the anti-inflammatory effects of exercise with the redox status of immune cells is still poorly understood. With the aim to verify the overall beneficial effect of regular training on the immune system, we have examined the acute and short-term effect of a 5-day exercise program on the modulation of protein and lipid oxidation, antioxidants (i.e., superoxide dismutase-1 (SOD1) and superoxide dismutase-2 (SOD2), glutathione peroxide 1 (GPx1), thioredoxin reductase-1 (TrxR1), and catalase (CAT)), and heat shock protein expression (i.e., heat shock protein-70 (HSP70) and heat shock protein-27 (HSP27)), at both mRNA and protein levels, as well as the activation of the nuclear factor kappa light chain enhancer of activated B cells (NFκB) in peripheral blood mononuclear cells (PBMCs). Moreover, plasmatic markers of oxidative stress, inflammation, and stress response (i.e., protein carbonyl content, interleukin-6 (IL6), interleukin-8 (IL8), interleukin-10 (IL10), interleukin-17E (IL17E), interleukin-17F (IL17F), interleukin-21 (IL21), interleukin-22 (IL22), and interleukin-23 (IL23)) were analyzed in active untrained young adult subjects. Even in the absence of an increased amount of protein or lipid oxidation, we confirmed a PBMC upregulation of SOD1 (1.26 ± 0.07 fold change, p < 0.05), HSP70 (1.59 ± 0.28 fold change, p < 0.05), and HSP27 gene expression (1.49 ± 0.09 fold change, p < 0.05) after 3 hours from the first bout of exercise, followed by an increase in proteins' amount at 24 hours (SOD1, 1.80 ± 0.34 fold change; HSP70, 3.40 ± 0.58 fold change; and HSP27, 1.81 ± 0.20 fold change, p < 0.05) and return to basal levels after the 5 days of aerobic training. Indeed, the posttraining basal levels of oxidized molecules in plasma and PBMCs were statistically lower than the pretraining levels (carbonyl content, 0.50 ± 0.05 fold change, p < 0.01), paralleled by a lower expression of SOD2, Gpx1, and TrxR1, at mRNA (SOD2, 0.63 ± 0.06; GPx1, 0.69 ± 0.07; and TrxR1, 0.69 ± 0.12 fold change, p < 0.05) and protein (TrxR1, 0.49 ± 0.11 fold change, p < 0.05) levels. These results verified the existence of an early phase of redox adaptation to physical exercise already achievable after 5 days of moderate, regular aerobic training. More interestingly, this phenomenon was paralleled by the degree of NFκB activation in PBMCs and the decrease of plasmatic proinflammatory cytokines IL8, IL21, and IL22 in the posttraining period, suggesting an interconnected, short-term efficacy of aerobic exercise towards systemic oxidative stress and inflammation.


Asunto(s)
Antioxidantes/uso terapéutico , Biomarcadores/metabolismo , Proteínas de Choque Térmico/metabolismo , Inflamación/metabolismo , Adulto , Ejercicio Físico/fisiología , Voluntarios Sanos , Humanos , Masculino
9.
J Cell Mol Med ; 13(9B): 3195-208, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19228261

RESUMEN

Stem cells capable of generating neural differentiated cells are recognized by the expression of nestin and reside in specific regions of the brain, namely, hippocampus, subventricular zone and olfactory bulb. For other brain structures, such as leptomeninges, which contribute to the correct cortex development and functions, there is no evidence so far that they may contain stem/precursor cells. In this work, we show for the first time that nestin-positive cells are present in rat leptomeninges during development up to adulthood. The newly identified nestin-positive cells can be extracted and expanded in vitro both as neurospheres, displaying high similarity with subventricular zone-derived neural stem cells, and as homogeneous cell population with stem cell features. In vitro expanded stem cell population can differentiate with high efficiency into excitable cells with neuronal phenotype and morphology. Once injected into the adult brain, these cells survive and differentiate into neurons, thus showing that their neuronal differentiation potential is operational also in vivo. In conclusion, our data provide evidence that a specific population of immature cells endowed of neuronal differentiation potential is resident in the leptomeninges throughout the life. As leptomeninges cover the entire central nervous system, these findings could have relevant implications for studies on cortical development and for regenerative medicine applied to neurological disorders.


Asunto(s)
Regulación de la Expresión Génica , Meninges/metabolismo , Neuronas/metabolismo , Células Madre/citología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Calcio/metabolismo , Proliferación Celular , Masculino , Microscopía Fluorescente/métodos , Ratas , Ratas Sprague-Dawley , Regeneración
10.
Stem Cells ; 26(1): 279-89, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17962701

RESUMEN

Bone marrow (BM)-derived mesenchymal stem cells (MSCs) are multipotent, nonhemopoietic progenitors that also possess regulatory activity on immune effector cells through different mechanisms. We demonstrate that human BM-derived MSCs expressed high levels of Toll-like receptors (TLRs) 3 and 4, which are both functional, as shown by the ability of their ligands to induce nuclear factor kappaB (NF-kappaB) activity, as well as the production of interleukin (IL)-6, IL-8, and CXCL10. Of note, ligation of TLR3 and TLR4 on MSCs also inhibited the ability of these cells to suppress the proliferation of T cells, without influencing their immunophenotype or differentiation potential. The TLR triggering effects appeared to be related to the impairment of MSC signaling to Notch receptors in T cells. Indeed, MSCs expressed the Notch ligand Jagged-1, and TLR3 or TLR4 ligation resulted in its strong downregulation. Moreover, anti-Jagged-1 neutralizing antibody and N[N-(3,5-difluorophenacetyl-l-alanyl)]-S-phenylglycine t-butyl ester (DAPT), an inhibitor of Notch signaling, hampered the suppressive activity of MSCs on T-cell proliferation. These data suggest that TLR3 and TLR4 expression on MSCs may provide an effective mechanism to block the immunosuppressive activity of MSCs and therefore to restore an efficient T-cell response in the course of dangerous infections, such as those sustained by double-stranded RNA viruses or Gram-negative bacteria, respectively.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Madre Mesenquimatosas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Quimiocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Células Madre Mesenquimatosas/citología , Microscopía Confocal , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Transfus Med Hemother ; 35(3): 194-204, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-21547117

RESUMEN

SUMMARY: Mesenchymal stem cells (MSCs) and their stromal progeny may be considered powerful regulatory cells, a sort of dendritic cell counterpart, which influence all the main immune effectors and functional roles in vivo, as well as potential applications in the treatment of a number of human immunological diseases. By choosing MSC tissue origin, cell dose, administration route, and treatment schedule, all the potential side effects related to MSC use, including tumor growth enhancement, have to be well considered to maximize the benefits of MSC-depen-dent immune regulation without significant risks for the patients.

12.
J Matern Fetal Neonatal Med ; 20(2): 145-9, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17437213

RESUMEN

OBJECTIVE: Preeclampsia is considered as a multifactorial disorder with a genetic predisposition. Alterations in the endothelin-1 (ET-1) system are considered to take part in triggering the vasoconstriction seen in preeclampsia. METHODS: In order to investigate the possible association of the -231 G > A polymorphism in the endothelin-1 type A receptor gene (EDNRA), previously shown to be associated with other conditions characterized by vasospasm, we examined 77 Caucasian preeclamptic women and 67 matched controls including normotensive subjects without history of thromboembolic event, abnormalities in blood pressure, proteinuria, edema and preeclampsia. The genotype was assessed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) on genomic DNA extracted from blood samples. Case vs. control allele frequencies and genotype distributions were compared. RESULTS: No significant differences were found when considering both genotype (chi(2) = 0.58, p = 0.75) and allelic frequencies (chi(2) = 0.08, p = 0.77). Furthermore, no significant genotype-related difference was found in relation to clinical features, such as gestational age at onset, systolic and diastolic blood pressure, proteinuria on admission and delivery week. CONCLUSIONS: No association between the -231 G > A polymorphism in the EDNRA gene and preeclampsia as well as any correlation with the main clinical features of the disorder were found, thus excluding a role for this polymorphism in susceptibility to preeclampsia.


Asunto(s)
Polimorfismo de Longitud del Fragmento de Restricción , Preeclampsia/genética , Receptor de Endotelina A/genética , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Reacción en Cadena de la Polimerasa , Embarazo , Análisis de Secuencia de ADN
13.
Stem Cells Dev ; 20(4): 709-19, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20695752

RESUMEN

Bone marrow mesenchymal stromal cells (BM-MSCs) may survive and proliferate in the presence of cycling neoplastic cells. Exogenously administered MSCs are actively incorporated in the tumor as stromal fibroblasts, thus competing with the local mesenchymal cell precursors. For this reason, MSCs have been suggested as a suitable carrier for gene therapy strategies, as they can be genetically engineered with genes encoding for biologically active molecules that can inhibit tumor cell proliferation and enhance the antitumor immune response. We used BM-MSCs engineered with the murine interferon-alpha (IFN-α) gene (BM-MSCs/IFN-α) to assess in a mouse plasmacytoma model the efficacy of this approach toward neoplastic plasma cells. We found that IFN-α can be efficiently produced and delivered inside the tumor microenvironment. Subcutaneous multiple administration of BM-MSCs/IFN-α significantly hampered the tumor growth in vivo and prolonged the overall survival of mice. The antitumor effect was associated with enhanced apoptosis of tumor cells, reduction in microvessel density, and ischemic necrosis. By contrast, intravenous administration of BM-MSCs/IFN-α did not significantly modify the survival of mice, mainly as a consequence of an excessive entrapment of injected cells in the pulmonary vessels. In conclusion, BM-MSCs/IFN-α are effective in inhibiting neoplastic plasma cell growth; however, systemic administration of engineered MSCs needs to be improved to make this approach potentially suitable for the treatment of multiple myeloma.


Asunto(s)
Interferón-alfa/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Plasmacitoma/terapia , Animales , Apoptosis , Células de la Médula Ósea/citología , Línea Celular Tumoral , Supervivencia Celular , Técnicas de Cocultivo , Terapia Genética , Interferón-alfa/genética , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Neovascularización Patológica/terapia , Células Plasmáticas/patología , Plasmacitoma/irrigación sanguínea , Plasmacitoma/patología , Proteínas Recombinantes/metabolismo , Antígenos Thy-1/metabolismo , Trasplante Heterólogo , Carga Tumoral
14.
J Mol Endocrinol ; 44(5): 259-69, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20150327

RESUMEN

Heterotrimeric G proteins transduce the signals of the largest family of membrane receptors (G protein-coupled receptors, GPCRs) hence triggering the activation of a wide variety of physiological responses. G15 is a G protein characterized by a number of functional peculiarities that make its signaling exceptional: 1) it can couple a variety of Gs-, Gi/o-, and Gq-linked receptors to phospholipase C activation; 2) relatively to other G proteins, it is poorly affected by beta-arrestin-dependent desensitization, the general mechanism that regulates GPCR function and 3) at the protein level, its expression is only detected in highly specific cell types (hematopoietic and epithelial cells). G15 alpha-subunit displays unique structural and biochemical properties, and is phylogenetically the most recent and divergent component of the Galphaq/11 subfamily. All these aspects shed a mysterious light on G15 biological role, which remains substantially elusive. Thus, far, G15 signaling has been analyzed in the context of hematopoiesis. Here, we highlight observations supporting the view that G15 functions may extend further beyond the immune system. In addition, we describe puzzling aspects of G15 signaling that offer a novel perspective in the understanding of its physiological role.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/fisiología , Transducción de Señal , Animales , Subunidades alfa de la Proteína de Unión al GTP/genética , Hematopoyesis , Humanos , Filogenia , Receptores Acoplados a Proteínas G/metabolismo
15.
Exp Cell Res ; 314(3): 603-15, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18022619

RESUMEN

It has recently been shown that adipose tissue is an abundant and easily accessible source of stromal progenitor cells (ADSCs, adipose-derived stromal cells), resembling the mesenchymal stem cells (MSCs) obtained from adult bone marrow. However, the identification of these two lineages is still controversial and even the stem cell nature of ADSCs is doubted. In this study, we examined the "stemness" transcriptional profile of ADSCs and BM-MSCs, with two aims: (1) to compare their "stem cell molecular signature" and (2) to dissect their constitutive expression pattern for molecules involved in tissue development, homeostasis and repair. As well as several molecules involved in matrix remodeling and adult tissue angiogenesis and repair, we detected the expression of genes UTF-1, Nodal, and Snail2, which are known to be expressed by embryonic stem cells but have been never described in other stem lineages. In addition, for the first time we described the transcriptional profile of human BM-MSCs and ADSCs for the CD44 splice variants, which are determinant in cell trafficking during embryonic development, in adult tissue homeostasis and also in tumor dissemination. Thus, our findings strongly support a close relationship between ADSCs and BM-MSCs, suggest an unexpected similarity between MSCs and embryonic stem cells, and possibly support the potential therapeutic application of ADSCs.


Asunto(s)
Tejido Adiposo/metabolismo , Células de la Médula Ósea/metabolismo , Linaje de la Célula/genética , Expresión Génica/genética , Células Madre Mesenquimatosas/metabolismo , Células del Estroma/metabolismo , Tejido Adiposo/citología , Empalme Alternativo/genética , Células de la Médula Ósea/citología , Moléculas de Adhesión Celular/genética , Diferenciación Celular/genética , Células Cultivadas , Citocinas/genética , Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Humanos , Receptores de Hialuranos/genética , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica/genética , Proteína Nodal , Proteínas Nucleares/genética , Regeneración/genética , Factores de Transcripción de la Familia Snail , Células del Estroma/citología , Transactivadores/genética , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/genética
16.
Headache ; 46(3): 486-91, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16618267

RESUMEN

OBJECTIVE: To assess whether the biallelic -231 G>A polymorphism of the endothelin type A receptor (EDNRA) gene, previously shown to be a marker of increased risk for developing migraine, has a role in the susceptibility to primary pediatric headache. BACKGROUND: Several studies suggest that endothelin has a role in migraine. A recent association study has shown that the biallelic -231 G>A polymorphism of the EDNRA gene is associated to migraine in an elderly population. METHODS: A total of 126 consecutive unrelated pediatric patients affected by primary headache, classified according to the International Headache Society criteria in migraine (migraine with aura, n = 3; migraine without aura, n = 80), and tension-type headache (episodic tension-type headache, n = 36; chronic tension-type headache, n = 7) patients, were recruited to the study. Sixty-seven healthy blood donors were used as a control group. Genomic DNA was extracted from buccal swabs or blood samples and analyzed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) for the above-mentioned polymorphism. Allele and genotype frequencies for primary headache patients were analyzed in comparison with the control group. RESULTS: No significant differences were found in the distribution of the EDNRA -231 G>A polymorphic variant when considering both genotype (migraine chi2 = 2.78, P = .25; tension-type headache chi2 = 3.58, P = .17) and allelic frequencies (migraine chi2 = 1.48, P = .22; tension-type headache chi2 = 0.39, P = .56). Furthermore, no significant genotype-related difference was found in relation to clinical features, such as age at onset, frequency, and length of the attacks. CONCLUSIONS: Our study shows that the -231 G>A polymorphism in the EDNRA gene is neither associated with primary juvenile headache nor significantly correlated with main clinical features characteristic of the headache pathology in pediatric settings.


Asunto(s)
Cefalea/genética , Polimorfismo Genético , Receptor de Endotelina A/genética , Adolescente , Niño , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino
17.
Pharmacogenet Genomics ; 16(11): 817-24, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17047490

RESUMEN

OBJECTIVE: The objective of the present study was to evaluate whether germline methylenetetrahydrofolate reductase (MTHFR) C677T and A1298C polymorphisms as well as polymorphisms in the thymidylate synthase gene promoter, namely the variable number tandem repeat polymorphism (TS VNTR) and the intrarepeat G to C single nucleotide polymorphism (TS SNP), are predictive markers of tumor regression in rectal cancer patients following preoperative chemoradiotherapy. BASIC METHODS: Blood samples from 125 patients with primary adenocarcinoma of the mid-low rectum who received 5-fluorouracil-based chemotherapy and external beam radiotherapy (median dose 48.4 Gy), 125 patients (women n=45, men n=80; median age 60 years, range 31-79 years) were genotyped. Response to preoperative treatment was evaluated employing the Tumor Regression Grade criteria. On the basis of the pathologic response, patients were classified as responders (TRG 1-2, n=48) and non-responders (TRG 3-5, n=74). Three patients were excluded because of insufficient data. MAIN RESULTS: Among the polymorphic variants examined, the MTHFR 677T-1298A haplotype was, upon univariate analysis, the only variable found associated with tumor regression (P=0.004). Moreover, at multivariate analysis, the MTHFR 677T-1298A haplotype was an independent predictor of tumor regression. Patients not carrying the MTHFR 677T-1298A haplotype (odds ratio 0.29, 95% confidence interval 0.13-0.64, P=0.002) displayed a higher response rate than patients with the MTHFR 677T-1298A haplotype. CONCLUSIONS: Unlike TS VNTR and SNP polymorphisms, MTHFR 677T-1298A haplotype in genomic DNA has the potential to be a predictive marker of tumor response in rectal cancer patients submitted to preoperative chemoradiotherapy.


Asunto(s)
Adenocarcinoma/diagnóstico , Haplotipos , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Neoplasias del Recto/diagnóstico , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Adenocarcinoma/radioterapia , Adenocarcinoma/cirugía , Adulto , Anciano , Antimetabolitos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica , Quimioterapia Adyuvante , Terapia Combinada , Femenino , Fluorouracilo/uso terapéutico , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Polimorfismo Genético , Pronóstico , Dosis de Radiación , Radioterapia Adyuvante , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/patología , Neoplasias del Recto/radioterapia , Neoplasias del Recto/cirugía , Inducción de Remisión
18.
J Headache Pain ; 6(4): 179-81, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16362657

RESUMEN

Numerous candidate genes for migraine have been proposed on the basis of their possible functional role in its pathogenesis. Genetic polymorphisms have been evaluated in association studies, some of which have been suggested to be susceptibility markers for adult migraine. To date, however, none of the identified polymorphisms in adult migraine susceptibility have been investigated in children, raising the possibility that they may not be necessarily involved in paediatric migraine susceptibility. This paper reviews studies of the genetic basis of migraine and summarises our experience in genetic association studies in primary paediatric headache susceptibility.


Asunto(s)
Trastornos Migrañosos/epidemiología , Trastornos Migrañosos/genética , Adulto , Distribución por Edad , Niño , Predisposición Genética a la Enfermedad/epidemiología , Humanos , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA