Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Res ; 17(7): 1531-1544, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30885992

RESUMEN

Hypoxia-inducible factor 1α is a key regulator of the hypoxia response in normal and cancer tissues. It is well recognized to regulate glycolysis and is a target for therapy. However, how tumor cells adapt to grow in the absence of HIF1α is poorly understood and an important concept to understand for developing targeted therapies is the flexibility of the metabolic response to hypoxia via alternative pathways. We analyzed pathways that allow cells to survive hypoxic stress in the absence of HIF1α, using the HCT116 colon cancer cell line with deleted HIF1α versus control. Spheroids were used to provide a 3D model of metabolic gradients. We conducted a metabolomic, transcriptomic, and proteomic analysis and integrated the results. These showed surprisingly that in three-dimensional growth, a key regulatory step of glycolysis is Aldolase A rather than phosphofructokinase. Furthermore, glucose uptake could be maintained in hypoxia through upregulation of GLUT14, not previously recognized in this role. Finally, there was a marked adaptation and change of phosphocreatine energy pathways, which made the cells susceptible to inhibition of creatine metabolism in hypoxic conditions. Overall, our studies show a complex adaptation to hypoxia that can bypass HIF1α, but it is targetable and it provides new insight into the key metabolic pathways involved in cancer growth. IMPLICATIONS: Under hypoxia and HIF1 blockade, cancer cells adapt their energy metabolism via upregulation of the GLUT14 glucose transporter and creatine metabolism providing new avenues for drug targeting.


Asunto(s)
Neoplasias del Colon/genética , Metabolismo Energético/genética , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias del Colon/patología , Creatina/genética , Creatina/metabolismo , Fructosa-Bifosfato Aldolasa/genética , Glucosa/metabolismo , Glucólisis/genética , Células HCT116 , Humanos , Esferoides Celulares/metabolismo , Hipoxia Tumoral/genética
2.
Nat Commun ; 8(1): 1258, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29097735

RESUMEN

AMPK is a conserved serine/threonine kinase whose activity maintains cellular energy homeostasis. Eukaryotic AMPK exists as αßγ complexes, whose regulatory γ subunit confers energy sensor function by binding adenine nucleotides. Humans bearing activating mutations in the γ2 subunit exhibit a phenotype including unexplained slowing of heart rate (bradycardia). Here, we show that γ2 AMPK activation downregulates fundamental sinoatrial cell pacemaker mechanisms to lower heart rate, including sarcolemmal hyperpolarization-activated current (I f) and ryanodine receptor-derived diastolic local subsarcolemmal Ca2+ release. In contrast, loss of γ2 AMPK induces a reciprocal phenotype of increased heart rate, and prevents the adaptive intrinsic bradycardia of endurance training. Our results reveal that in mammals, for which heart rate is a key determinant of cardiac energy demand, AMPK functions in an organ-specific manner to maintain cardiac energy homeostasis and determines cardiac physiological adaptation to exercise by modulating intrinsic sinoatrial cell behavior.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Bradicardia/genética , Calcio/metabolismo , Frecuencia Cardíaca/genética , Sarcolema/metabolismo , Nodo Sinoatrial/metabolismo , Adulto , Animales , Bradicardia/metabolismo , Electrocardiografía Ambulatoria , Ejercicio Físico , Corazón/diagnóstico por imagen , Humanos , Imagen por Resonancia Cinemagnética , Espectroscopía de Resonancia Magnética , Ratones , Microscopía Electrónica de Transmisión , Mutación , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Condicionamiento Físico Animal , Resistencia Física , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Nodo Sinoatrial/patología
3.
Oncotarget ; 8(40): 67439-67456, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978044

RESUMEN

Immunotherapy for metastatic melanoma offers great promise but, to date, only a subset of patients have responded. There is an urgent need to identify ways of allocating patients to the most beneficial therapy, to increase survival and decrease therapy-associated morbidity and costs. Blood-based biomarkers are of particular interest because of their straightforward implementation in routine clinical care. We sought to identify markers for dendritic cell (DC) vaccine-based immunotherapy against metastatic melanoma through gene expression analysis of peripheral blood mononuclear cells. A large-scale microarray analysis of 74 samples from two treatment centers, taken directly after the first round of DC vaccination, was performed. We found that phosphatidylethanolamine binding protein 1 (PEBP1)/Raf Kinase inhibitory protein (RKIP) expression can be used to identify a significant proportion of patients who performed poorly after DC vaccination. This result was validated by q-PCR analysis on blood samples from a second cohort of 95 patients treated with DC vaccination in four different centers. We conclude that low PEBP1 expression correlates with poor overall survival after DC vaccination. Intriguingly, this was only the case for expression of PEBP1 after, but not prior to, DC vaccination. Moreover, the change in PEBP1 expression upon vaccination correlated well with survival. Further analyses revealed that PEBP1 expression positively correlated with genes involved in T cell responses but inversely correlated with genes associated with myeloid cells and aberrant inflammation including STAT3, NOTCH1, and MAPK1. Concordantly, PEBP1 inversely correlated with the myeloid/lymphoid-ratio and was suppressed in patients suffering from chronic inflammatory disease.

4.
Wellcome Open Res ; 1: 17, 2016 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-28058287

RESUMEN

Background: Interleukin (IL)-27 is a member of the IL-6/IL-12 family of cytokines. It is a potent cytokine, with potential antiviral impact, and has been shown to play a role in modulating functions of diverse cell types, including Th1, Th2, and NK and B cells, demonstrating both pro- and anti-inflammatory roles.  In hepatocytes, it is capable of inducing signal transducer and activator of transcription (STAT)1, STAT3 and interferon-stimulated genes. Methods: To address its role in viral hepatitis, the antiviral activity of IL-27 against hepatitis C virus (HCV) and hepatitis B virus (HBV) was tested in vitro using cell-culture-derived infectious HCV (HCVcc) cell culture system and the HepaRG HBV cell culture model. To further investigate the impact of IL-27 on hepatocytes, Huh7.5 cells were treated with IL-27 to analyse the differentially expressed genes by microarray analysis. Furthermore, by quantitative PCR, we analyzed the up-regulation of chemokine (CXCL)-10 in response to IL-27. Results: In both HCV and HBV infection models, we observed only a modest direct antiviral effect. Microarray analysis showed that the up-regulated genes mostly belonged to antigen presentation and DNA replication pathways, and involved strong up-regulation of CXCL-10, a gene associated with liver inflammation. Overall, gene set enrichment analysis showed a striking correlation of these genes with those up-regulated in response to related cytokines in diverse cell populations. Conclusion: Our data indicate that IL-27 can have a significant pro-inflammatory impact in vitro, although the direct antiviral effect is modest. It may have a potential impact on hepatocyte function, especially chemokine expression and antigen presentation.

5.
Eur J Immunol ; 46(1): 167-77, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26460926

RESUMEN

MicroRNAs are important posttranscriptional regulators of gene expression, which have been shown to fine-tune innate immune responses downstream of pattern recognition receptor (PRR) signaling. This study identifies miR-650 as a novel PRR-responsive microRNA that is downregulated upon stimulation of primary human monocyte-derived dendritic cells (MDDCs) with a variety of different microbe-associated molecular patterns. A comprehensive target search combining in silico analysis, transcriptional profiling, and reporter assays reveals that miR-650 regulates several well-known interferon-stimulated genes, including IFIT2 and MXA. In particular, downregulation of miR-650 in influenza A infected MDDCs enhances the expression of MxA and may therefore contribute to the establishment of an antiviral state. Together these findings reveal a novel link between miR-650 and the innate immune response in human MDDCs.


Asunto(s)
Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Virus de la Influenza A/inmunología , MicroARNs/inmunología , Proteínas de Resistencia a Mixovirus/biosíntesis , Células Cultivadas , Células Dendríticas/metabolismo , Regulación hacia Abajo , Citometría de Flujo , Regulación de la Expresión Génica/genética , Humanos , Inmunidad Innata/inmunología , Immunoblotting , MicroARNs/genética , Microscopía Confocal , Proteínas de Resistencia a Mixovirus/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Reconocimiento de Patrones/inmunología , Transducción de Señal , Transfección
6.
Proc Natl Acad Sci U S A ; 112(49): 15172-7, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26598706

RESUMEN

A majority of breast cancers are driven by estrogen via estrogen receptor-α (ERα). Our previous studies indicate that hypoxia-inducible factor 1α (HIF-1α) cooperates with ERα in breast cancer cells. However, whether ERα is implicated in the direct regulation of HIF-1α and the role of HIF-1α in endocrine therapy response are unknown. In this study we found that a subpopulation of HIF-1α targets, many of them bearing both hypoxia response elements and estrogen response elements, are regulated by ERα in normoxia and hypoxia. Interestingly, the HIF-1α gene itself also bears an estrogen response element, and its expression is directly regulated by ERα. Clinical data revealed that expression of the HIF-1α gene or a hypoxia metagene signature is associated with a poor outcome to endocrine treatment in ERα(+) breast cancer. HIF-1α was able to confer endocrine therapy resistance to ERα(+) breast cancer cells. Our findings define, for the first time to our knowledge, a direct regulatory pathway between ERα and HIF-1α, which might modulate hormone response in treatment.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Moduladores de los Receptores de Estrógeno/uso terapéutico , Receptor alfa de Estrógeno/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Transducción de Señal , Tamoxifeno/uso terapéutico , Transcripción Genética/fisiología
7.
Cell Rep ; 13(8): 1578-88, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26586434

RESUMEN

Following exposure to vaccines, antigen-specific CD8(+) T cell responses develop as long-term memory pools. Vaccine strategies based on adenoviral vectors, e.g., those developed for HCV, are able to induce and sustain substantial CD8(+) T cell populations. How such populations evolve following vaccination remains to be defined at a transcriptional level. We addressed the transcriptional regulation of divergent CD8(+) T cell memory pools induced by an adenovector encoding a model antigen (beta-galactosidase). We observe transcriptional profiles that mimic those following infection with persistent pathogens, murine and human cytomegalovirus (CMV). Key transcriptional hallmarks include upregulation of homing receptors and anti-apoptotic pathways, driven by conserved networks of transcription factors, including T-bet. In humans, an adenovirus vaccine induced similar CMV-like phenotypes and transcription factor regulation. These data clarify the core features of CD8(+) T cell memory following vaccination with adenovectors and indicate a conserved pathway for memory development shared with persistent herpesviruses.


Asunto(s)
Adenoviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Vectores Genéticos/inmunología , Memoria Inmunológica/inmunología , Animales , Apoptosis/inmunología , Citomegalovirus/inmunología , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/inmunología , Vacunación/métodos
8.
Oncotarget ; 6(23): 19413-27, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26305601

RESUMEN

Carbonic anhydrase IX (CAIX) is strongly induced by hypoxia and its overexpression is associated with poor therapeutic outcome in cancer. Here, we report that hypoxia promotes tumour heterogeneity through the epigenetic regulation of CAIX. Based on hypoxic CAIX expression we identify and characterize two distinct populations of tumour cells, one that has inducible expression of CAIX and one that does not. The CAIX+ve population is enriched with cells expressing cancer stem cell markers and which have high self-renewal capacity. We show that differential CAIX expression is due to differences in chromatin structure. To further investigate the relationship between chromatin organization and hypoxic induction of CAIX expression we investigated the effect of JQ1 an inhibitor of BET bromodomain proteins and A366 a selective inhibitor of the H3K9 methyltransferase G9a/GLP. We identified that these drugs were able to modulate hypoxic CAIX expression induction. This further highlights the role of epigenetic modification in adaption to hypoxia and also in regulation of heterogeneity of cells within tumours. Interestingly, we identified that the two subpopulations show a differential sensitivity to HDAC inhibitors, NaBu or SAHA, with the CAIX positive showing greater sensitivity to treatment. We propose that drugs modulating chromatin regulation of expression may be used to reduce heterogeneity induced by hypoxia and could in combination have significant clinical consequences.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Anhidrasas Carbónicas/biosíntesis , Inhibidores de Histona Desacetilasas/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Animales , Antígenos de Neoplasias/genética , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/genética , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Inducción Enzimática , Femenino , Células HCT116 , Xenoinjertos , Humanos , Isoenzimas/biosíntesis , Células MCF-7 , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/patología
9.
Cell Rep ; 9(3): 1075-88, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25437561

RESUMEN

The C-type lectin CD161 is expressed by a large proportion of human T lymphocytes of all lineages, including a population known as mucosal-associated invariant T (MAIT) cells. To understand whether different T cell subsets expressing CD161 have similar properties, we examined these populations in parallel using mass cytometry and mRNA microarray approaches. The analysis identified a conserved CD161++/MAIT cell transcriptional signature enriched in CD161+CD8+ T cells, which can be extended to CD161+ CD4+ and CD161+TCRγδ+ T cells. Furthermore, this led to the identification of a shared innate-like, TCR-independent response to interleukin (IL)-12 plus IL-18 by different CD161-expressing T cell populations. This response was independent of regulation by CD161, which acted as a costimulatory molecule in the context of T cell receptor stimulation. Expression of CD161 hence identifies a transcriptional and functional phenotype, shared across human T lymphocytes and independent of both T cell receptor (TCR) expression and cell lineage.


Asunto(s)
Linaje de la Célula , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Transcripción Genética , Adulto , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/inmunología , Humanos , Interleucina-12/farmacología , Interleucina-18/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Fenotipo , Análisis de Componente Principal , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
10.
BMC Genomics ; 15: 224, 2014 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-24655320

RESUMEN

BACKGROUND: Oligonucleotide microarray-based comparative genomic hybridization (CGH) offers an attractive possible route for the rapid and cost-effective genome-wide discovery of deletion mutations. CGH typically involves comparison of the hybridization intensities of genomic DNA samples with microarray chip representations of entire genomes, and has widespread potential application in experimental research and medical diagnostics. However, the power to detect small deletions is low. RESULTS: Here we use a graduated series of Arabidopsis thaliana genomic deletion mutations (of sizes ranging from 4 bp to ~5 kb) to optimize CGH-based genomic deletion detection. We show that the power to detect smaller deletions (4, 28 and 104 bp) depends upon oligonucleotide density (essentially the number of genome-representative oligonucleotides on the microarray chip), and determine the oligonucleotide spacings necessary to guarantee detection of deletions of specified size. CONCLUSIONS: Our findings will enhance a wide range of research and clinical applications, and in particular will aid in the discovery of genomic deletions in the absence of a priori knowledge of their existence.


Asunto(s)
Hibridación Genómica Comparativa , Eliminación de Secuencia/genética , Arabidopsis/genética , ADN de Plantas/análisis , ADN de Plantas/metabolismo , Genoma de Planta , Análisis de Secuencia por Matrices de Oligonucleótidos
11.
Genom Data ; 2: 53-54, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26258045

RESUMEN

We performed array comparative genome hybridization (aCGH) analyses of five Arabidopsis thaliana mutants with genomic deletions ranging in size from 4 bp to > 5 kb. We used the Roche NimbleGen Arabidopsis CGH 3 × 720 K whole genome custom tiling array to optimize deletion detection. Details of the microarray design and hybridization data have been deposited at the NCBI GEO repository with accession number GSE55327.

12.
Mucosal Immunol ; 7(4): 763-774, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24280937

RESUMEN

Polymorphisms in the intracellular pattern recognition receptor gene NLRP3 (NLR family, pyrin domain containing 3) have been associated with susceptibility to Crohn's disease, a type of inflammatory bowel disease. Following tissue damage or infection, NLRP3 triggers the formation of inflammasomes, containing NLRP3, ASC (apoptosis-associated speck-like protein containing a CARD domain), and caspase-1, that mediate secretion of interleukin (IL)-1ß and IL-18. However, the precise role of NLRP3 inflammasomes in mucosal inflammation and barrier protection remains unclear. Here we show that upon infection with the attaching/effacing intestinal pathogen Citrobacter rodentium, Nlrp3(-/-) and Asc(-/-) mice displayed increased bacterial colonization and dispersion, more severe weight loss, and exacerbated intestinal inflammation. Analyses of irradiation bone marrow chimeras revealed that protection from disease was mediated through Nlrp3 activation in nonhematopoietic cells and was initiated very early after infection. Thus, early activation of Nlrp3 in intestinal epithelial cells limits pathogen colonization and prevents subsequent pathology, potentially providing a functional link between NLRP3 polymorphisms and susceptibility to inflammatory bowel disease.


Asunto(s)
Proteínas Portadoras/genética , Resistencia a la Enfermedad/genética , Interacciones Huésped-Patógeno/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Activación Transcripcional , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Citrobacter rodentium , Modelos Animales de Enfermedad , Resistencia a la Enfermedad/inmunología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Interacciones Huésped-Patógeno/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/microbiología , Mucosa Intestinal/microbiología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal
13.
Cell Rep ; 3(5): 1440-8, 2013 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-23643539

RESUMEN

The identification of mutated metabolic enzymes in hereditary cancer syndromes has established a direct link between metabolic dysregulation and cancer. Mutations in the Krebs cycle enzyme, fumarate hydratase (FH), predispose affected individuals to leiomyomas, renal cysts, and cancers, though the respective pathogenic roles of mitochondrial and cytosolic FH isoforms remain undefined. On the basis of comprehensive metabolomic analyses, we demonstrate that FH1-deficient cells and tissues exhibit defects in the urea cycle/arginine metabolism. Remarkably, transgenic re-expression of cytosolic FH ameliorated both renal cyst development and urea cycle defects associated with renal-specific FH1 deletion in mice. Furthermore, acute arginine depletion significantly reduced the viability of FH1-deficient cells in comparison to controls. Our findings highlight the importance of extramitochondrial metabolic pathways in FH-associated oncogenesis and the urea cycle/arginine metabolism as a potential therapeutic target.


Asunto(s)
Fumarato Hidratasa/metabolismo , Neoplasias Renales/enzimología , Animales , Arginina/metabolismo , Ácido Argininosuccínico/metabolismo , Línea Celular , Ciclo del Ácido Cítrico , Fumarato Hidratasa/deficiencia , Fumarato Hidratasa/genética , Fumaratos/metabolismo , Riñón/enzimología , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Metaboloma , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/metabolismo , Mutación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Urea/metabolismo
14.
Circ Res ; 112(11): 1433-43, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23529184

RESUMEN

RATIONALE: Smooth muscle cells (SMCs) are a key component of tissue-engineered vessels. However, the sources by which they can be isolated are limited. OBJECTIVE: We hypothesized that a large number of SMCs could be obtained by direct reprogramming of fibroblasts, that is, direct differentiation of specific cell lineages before the cells reaching the pluripotent state. METHODS AND RESULTS: We designed a combined protocol of reprogramming and differentiation of human neonatal lung fibroblasts. Four reprogramming factors (OCT4, SOX2, KLF4, and cMYC) were overexpressed in fibroblasts under reprogramming conditions for 4 days with cells defined as partially-induced pluripotent stem (PiPS) cells. PiPS cells did not form tumors in vivo after subcutaneous transplantation in severe combined immunodeficiency mice and differentiated into SMCs when seeded on collagen IV and maintained in differentiation media. PiPS-SMCs expressed a panel of SMC markers at mRNA and protein levels. Furthermore, the gene dickkopf 3 was found to be involved in the mechanism of PiPS-SMC differentiation. It was revealed that dickkopf 3 transcriptionally regulated SM22 by potentiation of Wnt signaling and interaction with Kremen1. Finally, PiPS-SMCs repopulated decellularized vessel grafts and ultimately gave rise to functional tissue-engineered vessels when combined with previously established PiPS-endothelial cells, leading to increased survival of severe combined immunodeficiency mice after transplantation of the vessel as a vascular graft. CONCLUSIONS: We developed a protocol to generate SMCs from PiPS cells through a dickkopf 3 signaling pathway, useful for generating tissue-engineered vessels. These findings provide a new insight into the mechanisms of SMC differentiation with vast therapeutic potential.


Asunto(s)
Prótesis Vascular , Fibroblastos/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pulmón/citología , Miocitos del Músculo Liso/citología , Células Madre Pluripotentes/citología , Proteínas Adaptadoras Transductoras de Señales , Diferenciación Celular/fisiología , Núcleo Celular/metabolismo , Separación Celular/métodos , Quimiocinas , Feto/citología , Fibroblastos/metabolismo , Humanos , Factor 4 Similar a Kruppel , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocitos del Músculo Liso/metabolismo , Activación Transcripcional/fisiología , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
15.
Proc Natl Acad Sci U S A ; 109(34): 13793-8, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869753

RESUMEN

The generation of induced pluripotent stem (iPS) cells is an important tool for regenerative medicine. However, the main restriction is the risk of tumor development. In this study we found that during the early stages of somatic cell reprogramming toward a pluripotent state, specific gene expression patterns are altered. Therefore, we developed a method to generate partial-iPS (PiPS) cells by transferring four reprogramming factors (OCT4, SOX2, KLF4, and c-MYC) to human fibroblasts for 4 d. PiPS cells did not form tumors in vivo and clearly displayed the potential to differentiate into endothelial cells (ECs) in response to defined media and culture conditions. To clarify the mechanism of PiPS cell differentiation into ECs, SET translocation (myeloid leukemia-associated) (SET) similar protein (SETSIP) was indentified to be induced during somatic cell reprogramming. Importantly, when PiPS cells were treated with VEGF, SETSIP was translocated to the cell nucleus, directly bound to the VE-cadherin promoter, increasing vascular endothelial-cadherin (VE-cadherin) expression levels and EC differentiation. Functionally, PiPS-ECs improved neovascularization and blood flow recovery in a hindlimb ischemic model. Furthermore, PiPS-ECs displayed good attachment, stabilization, patency, and typical vascular structure when seeded on decellularized vessel scaffolds. These findings indicate that reprogramming of fibroblasts into ECs via SETSIP and VEGF has a potential clinical application.


Asunto(s)
Reprogramación Celular , Células Endoteliales/citología , Fibroblastos/metabolismo , Neovascularización Patológica , Ingeniería de Tejidos/métodos , Animales , Antígenos CD/genética , Aorta/patología , Cadherinas/genética , Diferenciación Celular , Células Cultivadas , Fibroblastos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Factor 4 Similar a Kruppel , Ratones , Ratones SCID , Modelos Genéticos , Regiones Promotoras Genéticas , Células Madre/citología , Estrés Mecánico
16.
PLoS One ; 7(7): e39871, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22802946

RESUMEN

A major therapeutic challenge is how to replace bone once it is lost. Bone loss is a characteristic of chronic inflammatory and degenerative diseases such as rheumatoid arthritis and osteoporosis. Cells and cytokines of the immune system are known to regulate bone turnover by controlling the differentiation and activity of osteoclasts, the bone resorbing cells. However, less is known about the regulation of osteoblasts (OB), the bone forming cells. This study aimed to investigate whether immune cells also regulate OB differentiation. Using in vitro cell cultures of human bone marrow-derived mesenchymal stem cells (MSC), it was shown that monocytes/macrophages potently induced MSC differentiation into OBs. This was evident by increased alkaline phosphatase (ALP) after 7 days and the formation of mineralised bone nodules at 21 days. This monocyte-induced osteogenic effect was mediated by cell contact with MSCs leading to the production of soluble factor(s) by the monocytes. As a consequence of these interactions we observed a rapid activation of STAT3 in the MSCs. Gene profiling of STAT3 constitutively active (STAT3C) infected MSCs using Illumina whole human genome arrays showed that Runx2 and ALP were up-regulated whilst DKK1 was down-regulated in response to STAT3 signalling. STAT3C also led to the up-regulation of the oncostatin M (OSM) and LIF receptors. In the co-cultures, OSM that was produced by monocytes activated STAT3 in MSCs, and neutralising antibodies to OSM reduced ALP by 50%. These data indicate that OSM, in conjunction with other mediators, can drive MSC differentiation into OB. This study establishes a role for monocyte/macrophages as critical regulators of osteogenic differentiation via OSM production and the induction of STAT3 signalling in MSCs. Inducing the local activation of STAT3 in bone cells may be a valuable tool to increase bone formation in osteoporosis and arthritis, and in localised bone remodelling during fracture repair.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Monocitos/metabolismo , Osteoblastos/citología , Osteogénesis , Factor de Transcripción STAT3/fisiología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Humanos , Oncostatina M/fisiología , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
17.
Cancer Cell ; 20(4): 524-37, 2011 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-22014577

RESUMEN

The Krebs cycle enzyme fumarate hydratase (FH) is a human tumor suppressor whose inactivation is associated with the development of leiomyomata, renal cysts, and tumors. It has been proposed that activation of hypoxia inducible factor (HIF) by fumarate-mediated inhibition of HIF prolyl hydroxylases drives oncogenesis. Using a mouse model, we provide genetic evidence that Fh1-associated cyst formation is Hif independent, as is striking upregulation of antioxidant signaling pathways revealed by gene expression profiling. Mechanistic analysis revealed that fumarate modifies cysteine residues within the Kelch-like ECH-associated protein 1 (KEAP1), abrogating its ability to repress the Nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated antioxidant response pathway, suggesting a role for Nrf2 dysregulation in FH-associated cysts and tumors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Fumarato Hidratasa/fisiología , Fumaratos/metabolismo , Enfermedades Renales Quísticas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Succinatos/metabolismo , Animales , Antioxidantes/metabolismo , Hipoxia de la Célula , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Enfermedades Renales Quísticas/genética , Ratones , Procolágeno-Prolina Dioxigenasa/metabolismo , Transducción de Señal
18.
PLoS Biol ; 9(6): e1001086, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21738444

RESUMEN

The basic unit of genome packaging is the nucleosome, and nucleosomes have long been proposed to restrict DNA accessibility both to damage and to transcription. Nucleosome number in cells was considered fixed, but recently aging yeast and mammalian cells were shown to contain fewer nucleosomes. We show here that mammalian cells lacking High Mobility Group Box 1 protein (HMGB1) contain a reduced amount of core, linker, and variant histones, and a correspondingly reduced number of nucleosomes, possibly because HMGB1 facilitates nucleosome assembly. Yeast nhp6 mutants lacking Nhp6a and -b proteins, which are related to HMGB1, also have a reduced amount of histones and fewer nucleosomes. Nucleosome limitation in both mammalian and yeast cells increases the sensitivity of DNA to damage, increases transcription globally, and affects the relative expression of about 10% of genes. In yeast nhp6 cells the loss of more than one nucleosome in four does not affect the location of nucleosomes and their spacing, but nucleosomal occupancy. The decrease in nucleosomal occupancy is non-uniform and can be modelled assuming that different nucleosomal sites compete for available histones. Sites with a high propensity to occupation are almost always packaged into nucleosomes both in wild type and nucleosome-depleted cells; nucleosomes on sites with low propensity to occupation are disproportionately lost in nucleosome-depleted cells. We suggest that variation in nucleosome number, by affecting nucleosomal occupancy both genomewide and gene-specifically, constitutes a novel layer of epigenetic regulation.


Asunto(s)
Genoma , Proteína HMGB1/metabolismo , Histonas/metabolismo , Nucleosomas/metabolismo , Transcripción Genética , Animales , ADN/genética , ADN/metabolismo , Daño del ADN , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/fisiología , Proteína HMGB1/genética , Células HeLa , Histonas/genética , Humanos , Ratones , Modelos Teóricos , ARN/genética , ARN/metabolismo , Levaduras/genética , Levaduras/metabolismo
19.
PLoS Genet ; 7(7): e1002145, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21765815

RESUMEN

Forkhead-box protein P2 is a transcription factor that has been associated with intriguing aspects of cognitive function in humans, non-human mammals, and song-learning birds. Heterozygous mutations of the human FOXP2 gene cause a monogenic speech and language disorder. Reduced functional dosage of the mouse version (Foxp2) causes deficient cortico-striatal synaptic plasticity and impairs motor-skill learning. Moreover, the songbird orthologue appears critically important for vocal learning. Across diverse vertebrate species, this well-conserved transcription factor is highly expressed in the developing and adult central nervous system. Very little is known about the mechanisms regulated by Foxp2 during brain development. We used an integrated functional genomics strategy to robustly define Foxp2-dependent pathways, both direct and indirect targets, in the embryonic brain. Specifically, we performed genome-wide in vivo ChIP-chip screens for Foxp2-binding and thereby identified a set of 264 high-confidence neural targets under strict, empirically derived significance thresholds. The findings, coupled to expression profiling and in situ hybridization of brain tissue from wild-type and mutant mouse embryos, strongly highlighted gene networks linked to neurite development. We followed up our genomics data with functional experiments, showing that Foxp2 impacts on neurite outgrowth in primary neurons and in neuronal cell models. Our data indicate that Foxp2 modulates neuronal network formation, by directly and indirectly regulating mRNAs involved in the development and plasticity of neuronal connections.


Asunto(s)
Encéfalo/embriología , Factores de Transcripción Forkhead/genética , Redes Reguladoras de Genes , Neuritas/metabolismo , Proteínas Represoras/genética , Animales , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cuerpo Estriado/crecimiento & desarrollo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Cultivo Primario de Células , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
PLoS One ; 6(1): e16152, 2011 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-21264344

RESUMEN

MicroRNAs are small non-coding RNA molecules that regulate mRNA translation and stability by binding to complementary sequences usually within the 3' un-translated region (UTR). We have previously shown that the hepatic toxicity caused by wild-type Adenovirus 5 (Ad5WT) in mice can be prevented by incorporating 4 binding sites for the liver-specific microRNA, mir122, into the 3' UTR of E1A mRNA. This virus, termed Ad5mir122, is a promising virotherapy candidate and causes no obvious liver pathology. Herein we show that Ad5mir122 maintains wild-type lytic activity in cancer cells not expressing mir122 and assess any effects of possible mir122 depletion in host cells. Repeat administration of 2×10(10) viral particles of Admir122 to HepG2 tumour bearing mice showed significant anti-cancer efficacy. RT-QPCR showed that E1A mRNA was down-regulated 29-fold in liver when compared to Ad5WT. Western blot for E1A confirmed that all protein variants were knocked down. RT-QPCR for mature mir122 in infected livers showed that quantity of mir122 remained unaffected. Genome wide mRNA microarray profiling of infected livers showed that although the transcript level of >3900 different mRNAs changed more than 2-fold following Ad5WT infection, less than 600 were changed by Ad5mir122. These were then filtered to select mRNAs that were only altered by Ad5mir122 and the remaining 21 mRNAs were compared to predicted mir122 targets. No mir122 target mRNAs were affected by Ad5 mir122. These results demonstrate that the exploitation of microRNA regulation to control virus replication does not necessarily affect the level of the microRNA or the endogenous mRNA targets.


Asunto(s)
Adenoviridae/genética , MicroARNs/uso terapéutico , Neoplasias Experimentales/terapia , Animales , Células Hep G2 , Humanos , Hígado/química , Ratones , MicroARNs/análisis , Trasplante Heterólogo , Resultado del Tratamiento , Virión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...