Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Mol Cell ; 83(15): 2673-2691.e7, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37506700

RESUMEN

Cell cycle progression is linked to transcriptome dynamics and variations in the response of pluripotent cells to differentiation cues, mostly through unknown determinants. Here, we characterized the cell-cycle-associated transcriptome and proteome of mouse embryonic stem cells (mESCs) in naive ground state. We found that the thymine DNA glycosylase (TDG) is a cell-cycle-regulated co-factor of the tumor suppressor p53. Furthermore, TDG and p53 co-bind ESC-specific cis-regulatory elements and thereby control transcription of p53-dependent genes during self-renewal. We determined that the dynamic expression of TDG is required to promote the cell-cycle-associated transcriptional heterogeneity. Moreover, we demonstrated that transient depletion of TDG influences cell fate decisions during the early differentiation of mESCs. Our findings reveal an unanticipated role of TDG in promoting molecular heterogeneity during the cell cycle and highlight the central role of protein dynamics for the temporal control of cell fate during development.


Asunto(s)
Timina ADN Glicosilasa , Proteína p53 Supresora de Tumor , Animales , Ratones , Ciclo Celular/genética , Línea Celular , Regulación de la Expresión Génica , Timina ADN Glicosilasa/genética , Timina ADN Glicosilasa/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
J Clin Endocrinol Metab ; 107(1): 150-166, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34487152

RESUMEN

CONTEXT: Chronic glucocorticoid (GC) overexposure, resulting from endogenous Cushing's syndrome (CS) or exogenous GC therapy, causes several adverse outcomes, including persistent central fat accumulation associated with a low-grade inflammation. However, no previous multiomics studies in visceral adipose tissue (VAT) from patients exposed to high levels of unsuppressed GC during active CS or after remission are available yet. OBJECTIVE: To determine the persistent VAT transcriptomic alterations and epigenetic fingerprints induced by chronic hypercortisolism. METHODS: We employed a translational approach combining high-throughput data on endogenous CS patients and a reversible CS mouse model. We performed RNA sequencing and chromatin immunoprecipitation sequencing on histone modifications (H3K4me3, H3K27ac, and H3K27me3) to identify persistent transcriptional and epigenetic signatures in VAT produced during active CS and maintained after remission. RESULTS: VAT dysfunction was associated with low-grade proinflammatory status, macrophage infiltration, and extracellular matrix remodeling. Most notably, chronic hypercortisolism caused a persistent circadian rhythm disruption in VAT through core clock genes modulation. Importantly, changes in the levels of 2 histone modifications associated to gene transcriptional activation (H3K4me3 and H3K27ac) correlated with the observed differences in gene expression during active CS and after CS remission. CONCLUSION: We identified for the first time the persistent transcriptional and epigenetic signatures induced by hypercortisolism in VAT, providing a novel integrated view of molecular components driving the long-term VAT impairment associated with CS.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/complicaciones , Síndrome de Cushing/metabolismo , Glucocorticoides/efectos adversos , Grasa Intraabdominal/inmunología , Obesidad Abdominal/genética , Administración Oral , Neoplasias de las Glándulas Suprarrenales/diagnóstico , Neoplasias de las Glándulas Suprarrenales/inmunología , Neoplasias de las Glándulas Suprarrenales/orina , Adulto , Animales , Biopsia , Secuenciación de Inmunoprecipitación de Cromatina , Corticosterona/administración & dosificación , Corticosterona/efectos adversos , Estudios Transversales , Síndrome de Cushing/inmunología , Síndrome de Cushing/patología , Modelos Animales de Enfermedad , Epigenoma/efectos de los fármacos , Epigenoma/inmunología , Femenino , Glucocorticoides/administración & dosificación , Glucocorticoides/metabolismo , Humanos , Hidrocortisona/metabolismo , Hidrocortisona/orina , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Masculino , Ratones , Persona de Mediana Edad , Obesidad Abdominal/inmunología , Obesidad Abdominal/patología , RNA-Seq , Transcriptoma/efectos de los fármacos , Transcriptoma/inmunología
3.
Front Cell Dev Biol ; 9: 655201, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33996816

RESUMEN

Polycomb group (PcG) of proteins are a group of highly conserved epigenetic regulators involved in many biological functions, such as embryonic development, cell proliferation, and adult stem cell determination. PHD finger protein 19 (PHF19) is an associated factor of Polycomb repressor complex 2 (PRC2), often upregulated in human cancers. In particular, myeloid leukemia cell lines show increased levels of PHF19, yet little is known about its function. Here, we have characterized the role of PHF19 in myeloid leukemia cells. We demonstrated that PHF19 depletion decreases cell proliferation and promotes chronic myeloid leukemia (CML) differentiation. Mechanistically, we have shown how PHF19 regulates the proliferation of CML through a direct regulation of the cell cycle inhibitor p21. Furthermore, we observed that MTF2, a PHF19 homolog, partially compensates for PHF19 depletion in a subset of target genes, instructing specific erythroid differentiation. Taken together, our results show that PHF19 is a key transcriptional regulator for cell fate determination and could be a potential therapeutic target for myeloid leukemia treatment.

4.
Front Cell Dev Biol ; 9: 654344, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33869213

RESUMEN

Adenosylhomocysteinase (AHCY) is a unique enzyme and one of the most conserved proteins in living organisms. AHCY catalyzes the reversible break of S-adenosylhomocysteine (SAH), the by-product and a potent inhibitor of methyltransferases activity. In mammals, AHCY is the only enzyme capable of performing this reaction. Controlled subcellular localization of AHCY is believed to facilitate local transmethylation reactions, by removing excess of SAH. Accordingly, AHCY is recruited to chromatin during replication and active transcription, correlating with increasing demands for DNA, RNA, and histone methylation. AHCY deletion is embryonic lethal in many organisms (from plants to mammals). In humans, AHCY deficiency is associated with an incurable rare recessive disorder in methionine metabolism. In this review, we focus on the AHCY protein from an evolutionary, biochemical, and functional point of view, and we discuss the most recent, relevant, and controversial contributions to the study of this enzyme.

5.
Sci Adv ; 6(32): eabb2745, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32821835

RESUMEN

Adult hematopoietic stem cells (HSCs) are rare multipotent cells in bone marrow that are responsible for generating all blood cell types. HSCs are a heterogeneous group of cells with high plasticity, in part, conferred by epigenetic mechanisms. PHF19, a subunit of the Polycomb repressive complex 2 (PRC2), is preferentially expressed in mouse hematopoietic precursors. Here, we now show that, in stark contrast to results published for other PRC2 subunits, genetic depletion of Phf19 increases HSC identity and quiescence. While proliferation of HSCs is normally triggered by forced mobilization, defects in differentiation impede long-term correct blood production, eventually leading to aberrant hematopoiesis. At molecular level, PHF19 deletion triggers a redistribution of the histone repressive mark H3K27me3, which notably accumulates at blood lineage-specific genes. Our results provide novel insights into how epigenetic mechanisms determine HSC identity, control differentiation, and are key for proper hematopoiesis.

6.
Elife ; 92020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32155117

RESUMEN

The Polycomb-like protein PHF19/PCL3 associates with PRC2 and mediates its recruitment to chromatin in embryonic stem cells. PHF19 is also overexpressed in many cancers. However, neither PHF19 targets nor misregulated pathways involving PHF19 are known. Here, we investigate the role of PHF19 in prostate cancer cells. We find that PHF19 interacts with PRC2 and binds to PRC2 targets on chromatin. PHF19 target genes are involved in proliferation, differentiation, angiogenesis, and extracellular matrix organization. Depletion of PHF19 triggers an increase in MTF2/PCL2 chromatin recruitment, with a genome-wide gain in PRC2 occupancy and H3K27me3 deposition. Transcriptome analysis shows that PHF19 loss promotes deregulation of key genes involved in growth, metastasis, invasion, and of factors that stimulate blood vessels formation. Consistent with this, PHF19 silencing reduces cell proliferation, while promotes invasive growth and angiogenesis. Our findings reveal a role for PHF19 in controlling the balance between cell proliferation and invasiveness in prostate cancer.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias de la Próstata/metabolismo , Factores de Transcripción/metabolismo , Fosfatos de Calcio/metabolismo , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Neoplasias de la Próstata/genética , Factores de Transcripción/genética
7.
Elife ; 72018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29376829

RESUMEN

The best characterized signaling pathway downstream of transforming growth factor ß (TGF-ß) is through SMAD2 and SMAD3. However, TGF-ß also induces phosphorylation of SMAD1 and SMAD5, but the mechanism of this phosphorylation and its functional relevance is not known. Here, we show that TGF-ß-induced SMAD1/5 phosphorylation requires members of two classes of type I receptor, TGFBR1 and ACVR1, and establish a new paradigm for receptor activation where TGFBR1 phosphorylates and activates ACVR1, which phosphorylates SMAD1/5. We demonstrate the biological significance of this pathway by showing that approximately a quarter of the TGF-ß-induced transcriptome depends on SMAD1/5 signaling, with major early transcriptional targets being the ID genes. Finally, we show that TGF-ß-induced epithelial-to-mesenchymal transition requires signaling via both the SMAD3 and SMAD1/5 pathways, with SMAD1/5 signaling being essential to induce ID1. Therefore, combinatorial signaling via both SMAD pathways is essential for the full TGF-ß-induced transcriptional program and physiological responses.


Asunto(s)
Transición Epitelial-Mesenquimal , Procesamiento Proteico-Postraduccional , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Activinas Tipo I/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Fosforilación , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo
8.
Front Mol Biosci ; 4: 8, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28321398

RESUMEN

Rhabdomyolysis is a disorder characterized by acute damage of the sarcolemma of the skeletal muscle leading to release of potentially toxic muscle cell components into the circulation, most notably creatine phosphokinase (CK) and myoglobulin, and is frequently accompanied by myoglobinuria. In the present work, we evaluated the toxicity of p-phenylenediamine (PPD), a main component of hair dyes which is reported to induce rhabdomyolysis. We studied the metabolic effect of this compound in vivo with Wistar rats and in vitro with C2C12 muscle cells. To this aim we have combined multi-omic experimental measurements with computational approaches using model-driven methods. The integrative study presented here has unveiled the metabolic disorders associated to PPD exposure that may underlay the aberrant metabolism observed in rhabdomyolys disease. Animals treated with lower doses of PPD (10 and 20 mg/kg) showed depressed activity and myoglobinuria after 10 h of treatment. We measured the serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and creatine kinase (CK) in rats after 24, 48, and 72 h of PPD exposure. At all times, treatment with PPD at higher doses (40 and 60 mg/kg) showed an increase of AST and ALT, and also an increase of lactate dehydrogenase (LDH) and CK after 24 h. Blood packed cell volume and hemoglobin levels, as well as organs weight at 48 and 72 h, were also measured. No significant differences were observed in these parameters under any condition. PPD induce cell cycle arrest in S phase and apoptosis (40% or early apoptotic cells) on mus musculus mouse C2C12 cells after 24 h of treatment. Incubation of mus musculus mouse C2C12 cells with [1,2-13C2]-glucose during 24 h, subsequent quantification of 13C isotopologues distribution in key metabolites of glucose metabolic network and a computational fluxomic analysis using in-house developed software (Isodyn) showed that PPD is inhibiting glycolysis, non-oxidative pentose phosphate pathway, glycogen turnover, and ATPAse reaction leading to a reduction in ATP synthesis. These findings unveil the glucose metabolism collapse, which is consistent with a decrease in cell viability observed in PPD-treated C2C12 cells and with the myoglubinuria and other effects observed in Wistar Rats treated with PPD. These findings shed new light on muscle dysfunction associated to PPD exposure, opening new avenues for cost-effective therapies in Rhabdomyolysis disease.

9.
Mol Cell ; 64(4): 645-658, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863225

RESUMEN

The cellular plasticity of pluripotent stem cells is thought to be sustained by genomic regions that display both active and repressive chromatin properties. These regions exhibit low levels of gene expression, yet the mechanisms controlling these levels remain unknown. Here, we describe Elongin BC as a binding factor at the promoters of bivalent sites. Biochemical and genome-wide analyses show that Elongin BC is associated with Polycomb Repressive Complex 2 (PRC2) in pluripotent stem cells. Elongin BC is recruited to chromatin by the PRC2-associated factor EPOP (Elongin BC and Polycomb Repressive Complex 2 Associated Protein, also termed C17orf96, esPRC2p48, E130012A19Rik), a protein expressed in the inner cell mass of the mouse blastocyst. Both EPOP and Elongin BC are required to maintain low levels of expression at PRC2 genomic targets. Our results indicate that keeping the balance between activating and repressive cues is a more general feature of chromatin in pluripotent stem cells than previously appreciated.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Células Madre Pluripotentes/metabolismo , Complejo Represivo Polycomb 2/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular , Cromatina/química , Cromatina/metabolismo , Proteínas Cromosómicas no Histona , ADN Polimerasa II/genética , ADN Polimerasa II/metabolismo , Elonguina , Implantación del Embrión , Embrión de Mamíferos , Histonas/genética , Histonas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células Madre Pluripotentes/citología , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción/metabolismo , Transcripción Genética
11.
Mol Cell Biol ; 36(5): 809-19, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26711262

RESUMEN

Discovering and classifying long noncoding RNAs (lncRNAs) across all mammalian tissues and cell lines remains a major challenge. Previously, mouse lncRNAs were identified using transcriptome sequencing (RNA-seq) data from a limited number of tissues or cell lines. Additionally, associating a few hundred lncRNA promoters with chromatin states in a single mouse cell line has identified two classes of chromatin-associated lncRNA. However, the discovery and classification of lncRNAs is still pending in many other tissues in mouse. To address this, we built a comprehensive catalog of lncRNAs by combining known lncRNAs with high-confidence novel lncRNAs identified by mapping and de novo assembling billions of RNA-seq reads from eight tissues and a primary cell line in mouse. Next, we integrated this catalog of lncRNAs with multiple genome-wide chromatin state maps and found two different classes of chromatin state-associated lncRNAs, including promoter-associated (plncRNAs) and enhancer-associated (elncRNAs) lncRNAs, across various tissues. Experimental knockdown of an elncRNA resulted in the downregulation of the neighboring protein-coding Kdm8 gene, encoding a histone demethylase. Our findings provide 2,803 novel lncRNAs and a comprehensive catalog of chromatin-associated lncRNAs across different tissues in mouse.


Asunto(s)
Cromatina/genética , Ratones/genética , ARN Largo no Codificante/genética , Animales , Mapeo Cromosómico , Perfilación de la Expresión Génica , Regiones Promotoras Genéticas , Análisis de Secuencia de ARN , Transcriptoma
12.
Nat Commun ; 6: 7264, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-26081042

RESUMEN

Sprouting angiogenesis drives blood vessel growth in healthy and diseased tissues. Vegf and Dll4/Notch signalling cooperate in a negative feedback loop that specifies endothelial tip and stalk cells to ensure adequate vessel branching and function. Current concepts posit that endothelial cells default to the tip-cell phenotype when Notch is inactive. Here we identify instead that the stalk-cell phenotype needs to be actively repressed to allow tip-cell formation. We show this is a key endothelial function of neuropilin-1 (Nrp1), which suppresses the stalk-cell phenotype by limiting Smad2/3 activation through Alk1 and Alk5. Notch downregulates Nrp1, thus relieving the inhibition of Alk1 and Alk5, thereby driving stalk-cell behaviour. Conceptually, our work shows that the heterogeneity between neighbouring endothelial cells established by the lateral feedback loop of Dll4/Notch utilizes Nrp1 levels as the pivot, which in turn establishes differential responsiveness to TGF-ß/BMP signalling.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Endotelio Vascular/crecimiento & desarrollo , Neuropilina-1/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Notch/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Receptores de Activinas Tipo II , Animales , Factor 2 de Diferenciación de Crecimiento/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Fenotipo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
13.
FEBS J ; 282(9): 1723-35, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25271128

RESUMEN

The Polycomb group (PcG) of proteins form chromatin-binding complexes with histone-modifying activity. The two main PcG repressive complexes studied (PRC1 and PRC2) are generally associated with chromatin in its repressed state. PRC2 is responsible for methylation of histone H3 at lysine 27 (H3K27me3), an epigenetic mark that is linked with numerous biological processes, including development, adult homeostasis and cancer. The core canonical complex PRC2, which contains the EZH1/2, SUZ12 and EED proteins, may be extended and functionally manipulated through interactions with several other proteins. In this review, we focus on these PRC2-associated proteins. As PRC2 functions are diverse, the variability conferred by these sub-stoichiometrically associated members may help to understand specific changes in PRC2 activity, chromatin recruitment and distribution required for gene repression.


Asunto(s)
Enfermedad , Proteínas del Grupo Polycomb/fisiología , Células Madre/citología , Animales , Humanos , Metilación , Proteínas del Grupo Polycomb/metabolismo , Células Madre/metabolismo
14.
Sci Signal ; 7(344): lc2, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25249656

RESUMEN

Using an integrative experimental and computational modeling approach to dissect the signaling dynamics of the transforming growth factor-ß to Smad (TGF-ß/Smad) pathway, we discovered that previous exposure to ligand desensitizes cells, rendering them refractory to further acute TGF-ß stimulation. We demonstrated that this refractory behavior, which also explains signal attenuation, is caused by the fast depletion from the cell surface of signaling-competent receptors upon TGF-ß binding and their slow replenishment, which is the rate-limiting step for regaining full competence for acute ligand induction. In their Comment, Warmflash and colleagues suggest that receptor dynamics do not necessarily reflect the dynamics of TGF-ß target gene transcription. We argue that to understand receptor dynamics, phosphorylated Smad2 abundance is the optimal readout, because this directly reflects receptor activity. Target gene transcription, in contrast, is influenced by many other factors in addition to nuclear abundance of activated Smad complexes and is thus a poor readout for receptor dynamics. Warmflash et al. also claim that our results are inconsistent with parts of the literature, in particular with data published by Zi et al. (Mol. Syst. Biol. 7, 492, 2011) and by Sorre et al. (Dev. Cell 20, 334, 2014). However, we show with our mathematical model that our results are consistent with the data in question.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Humanos
15.
Sci Signal ; 6(305): ra106, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24327760

RESUMEN

Understanding the complex dynamics of growth factor signaling requires both mechanistic and kinetic information. Although signaling dynamics have been studied for pathways downstream of receptor tyrosine kinases and G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors, they have not been investigated for the transforming growth factor-ß (TGF-ß) superfamily pathways. Using an integrative experimental and mathematical modeling approach, we dissected the dynamic behavior of the TGF-ß to Smad pathway, which is mediated by type I and type II receptor serine/threonine kinases, in response to acute, chronic, and repeated ligand stimulations. TGF-ß exposure produced a transient response that attenuated over time, resulting in desensitized cells that were refractory to further acute stimulation. This loss of signaling competence depended on ligand binding, but not on receptor activity, and was restored only after the ligand had been depleted. Furthermore, TGF-ß binding triggered the rapid depletion of signaling-competent receptors from the cell surface, with the type I and type II receptors exhibiting different degradation and trafficking kinetics. A computational model of TGF-ß signal transduction from the membrane to the nucleus that incorporates our experimental findings predicts that autocrine signaling, such as that associated with tumorigenesis, severely compromises the TGF-ß response, which we confirmed experimentally. Thus, we have shown that the long-term signaling behavior of the TGF-ß pathway is determined by receptor dynamics, does not require TGF-ß-induced gene expression, and influences context-dependent responses in vivo.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Western Blotting , Línea Celular , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Cinética , Ligandos , Modelos Biológicos , Fosforilación , Unión Proteica , Transporte de Proteínas/efectos de los fármacos , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
16.
Proc Natl Acad Sci U S A ; 110(45): 18250-5, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24145436

RESUMEN

Mantle cell lymphoma (MCL) is an aggressive tumor, but a subset of patients may follow an indolent clinical course. To understand the mechanisms underlying this biological heterogeneity, we performed whole-genome and/or whole-exome sequencing on 29 MCL cases and their respective matched normal DNA, as well as 6 MCL cell lines. Recurrently mutated genes were investigated by targeted sequencing in an independent cohort of 172 MCL patients. We identified 25 significantly mutated genes, including known drivers such as ataxia-telangectasia mutated (ATM), cyclin D1 (CCND1), and the tumor suppressor TP53; mutated genes encoding the anti-apoptotic protein BIRC3 and Toll-like receptor 2 (TLR2); and the chromatin modifiers WHSC1, MLL2, and MEF2B. We also found NOTCH2 mutations as an alternative phenomenon to NOTCH1 mutations in aggressive tumors with a dismal prognosis. Analysis of two simultaneous or subsequent MCL samples by whole-genome/whole-exome (n = 8) or targeted (n = 19) sequencing revealed subclonal heterogeneity at diagnosis in samples from different topographic sites and modulation of the initial mutational profile at the progression of the disease. Some mutations were predominantly clonal or subclonal, indicating an early or late event in tumor evolution, respectively. Our study identifies molecular mechanisms contributing to MCL pathogenesis and offers potential targets for therapeutic intervention.


Asunto(s)
Evolución Clonal/genética , Variación Genética , Genoma Humano/genética , Linfoma de Células del Manto/genética , Mutación/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Secuencia de Bases , Ciclina D1/genética , Estudio de Asociación del Genoma Completo , Genómica/métodos , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Linfoma de Células del Manto/fisiopatología , Análisis por Micromatrices , Datos de Secuencia Molecular , Receptor Notch2/genética , Receptor Toll-Like 2/genética
17.
Mol Cell Biol ; 32(14): 2904-16, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22615489

RESUMEN

In vivo cells receive simultaneous signals from multiple extracellular ligands and must integrate and interpret them to respond appropriately. Here we investigate the interplay between pathways downstream of two transforming growth factor ß (TGF-ß) superfamily members, bone morphogenetic protein (BMP) and TGF-ß. We show that in multiple cell lines, TGF-ß potently inhibits BMP-induced transcription at the level of both BMP-responsive reporter genes and endogenous BMP target genes. This inhibitory effect requires the TGF-ß type I receptor ALK5 and is independent of new protein synthesis. Strikingly, we show that Smad3 is required for TGF-ß's inhibitory effects, whereas Smad2 is not. We go on to demonstrate that TGF-ß induces the formation of complexes comprising phosphorylated Smad1/5 and Smad3, which bind to BMP-responsive elements in vitro and in vivo and mediate TGF-ß-induced transcriptional repression. Furthermore, loss of Smad3 confers on TGF-ß the ability to induce transcription via BMP-responsive elements. Our results therefore suggest that not only is Smad3 important for mediating TGF-ß's inhibitory effects on BMP signaling but it also plays a critical role in restricting the transcriptional output in response to TGF-ß.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteína Smad1/metabolismo , Proteína smad3/metabolismo , Proteína Smad5/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Secuencia de Bases , Proteína Morfogenética Ósea 7/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular , Línea Celular Tumoral , Cartilla de ADN/genética , Humanos , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Invasividad Neoplásica , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Smad1/química , Proteína Smad1/genética , Proteína smad3/química , Proteína smad3/genética , Proteína Smad4/química , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína Smad5/química , Proteína Smad5/genética , Transcripción Genética/efectos de los fármacos
18.
Biochim Biophys Acta ; 1807(6): 746-54, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21185256

RESUMEN

Metabolic adaptations are associated with changes in enzyme activities. These adaptations are characterized by patterns of positive and negative changes in metabolic fluxes and concentrations of intermediate metabolites. Knowledge of the mechanism and parameters governing enzyme kinetics is rarely available. However, the signs-increases or decreases-of many of these changes can be predicted using the signs of metabolic control coefficients. These signs require the only knowledge of the structure of the metabolic network and a limited qualitative knowledge of the regulatory dependences, which is widely available for carbon metabolism. Here, as a case study, we identified control coefficients with fixed signs in order to predict the pattern of changes in key enzyme activities which can explain the observed changes in fluxes and concentrations underlying the metabolic adaptations in oncogenic K-ras transformation in NIH-3T3 cells. The fixed signs of control coefficients indicate that metabolic changes following the oncogenic transformation-increased glycolysis and oxidative branch of the pentose-phosphate pathway, and decreased concentration in sugar-phosphates-could be associated with increases in activity for glucose-6-phosphate dehydrogenase, pyruvate kinase and lactate dehydrogenase, and decrease for transketolase. These predictions were validated experimentally by measuring specific activities. We conclude that predictions based on fixed signs of control coefficients are a very robust tool for the identification of changes in enzyme activities that can explain observed metabolic adaptations in carbon metabolism.


Asunto(s)
Adaptación Biológica , Carbono/metabolismo , Transformación Celular Neoplásica/genética , Genes ras , Redes y Vías Metabólicas , Adaptación Biológica/genética , Adaptación Biológica/fisiología , Animales , Transporte Biológico , Metabolismo de los Hidratos de Carbono/genética , Metabolismo de los Hidratos de Carbono/fisiología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Genes ras/fisiología , Redes y Vías Metabólicas/genética , Redes y Vías Metabólicas/fisiología , Ratones , Modelos Biológicos , Células 3T3 NIH , Concentración Osmolar , Transfección
19.
BMC Syst Biol ; 4: 135, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20925932

RESUMEN

BACKGROUND: Metabolic flux profiling based on the analysis of distribution of stable isotope tracer in metabolites is an important method widely used in cancer research to understand the regulation of cell metabolism and elaborate new therapeutic strategies. Recently, we developed software Isodyn, which extends the methodology of kinetic modeling to the analysis of isotopic isomer distribution for the evaluation of cellular metabolic flux profile under relevant conditions. This tool can be applied to reveal the metabolic effect of proapoptotic drug edelfosine in leukemia Jurkat cell line, uncovering the mechanisms of induction of apoptosis in cancer cells. RESULTS: The study of 13C distribution of Jukat cells exposed to low edelfosine concentration, which induces apoptosis in ≤5% of cells, revealed metabolic changes previous to the development of apoptotic program. Specifically, it was found that low dose of edelfosine stimulates the TCA cycle. These metabolic perturbations were coupled with an increase of nucleic acid synthesis de novo, which indicates acceleration of biosynthetic and reparative processes. The further increase of the TCA cycle fluxes, when higher doses of drug applied, eventually enhance reactive oxygen species (ROS) production and trigger apoptotic program. CONCLUSION: The application of Isodyn to the analysis of mechanism of edelfosine-induced apoptosis revealed primary drug-induced metabolic changes, which are important for the subsequent initiation of apoptotic program. Initiation of such metabolic changes could be exploited in anticancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Éteres Fosfolípidos/farmacología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Especies Reactivas de Oxígeno/metabolismo , Glucosa/metabolismo , Humanos , Isótopos , Células Jurkat , Ácido Láctico/biosíntesis
20.
Biochem Soc Trans ; 38(5): 1302-6, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20863303

RESUMEN

Metabolite concentrations and fluxes are the system variables that characterize metabolism. The systematic study of metabolite profiles is known as metabolomics; however, knowledge of the complete set of metabolites may not be enough to predict distinct phenotypes. A complete understanding of metabolic processes requires detailed knowledge of enzyme-controlled intracellular fluxes. These can be estimated through quantitative measurements of metabolites at different times or by analysing the stable isotope patterns obtained after incubation with labelled substrates. We have identified distinct intracellular fluxes associated with metabolic adaptations accompanying cancer. The maintenance of an imbalance between fluxes for the oxidative and non-oxidative PPP (pentose phosphate pathway) has been shown to be critical for angiogenesis and cancer cell survival. Mouse NIH 3T3 cells transformed by different mutated K-ras oncogenes have differential routing of glucose to anaerobic glycolysis, the PPP and the Krebs cycle. These results indicate that knowledge of metabolic fingerprints associated with an altered genetic profile could be exploited in the rational design of new therapies. We conclude that the understanding of the multifactorial nature of metabolic adaptations in cancer may open new ways to develop novel multi-hit antitumoral therapies.


Asunto(s)
Redes y Vías Metabólicas/fisiología , Neoplasias/metabolismo , Animales , Antineoplásicos/uso terapéutico , Humanos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Biología de Sistemas/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...