Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 171(4): 736-737, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100069

RESUMEN

Nearly 3% of the human population carries bi-allelic loss-of-function variants in the gene encoding CLYBL. While largely healthy, these individuals exhibit reduced circulating vitamin B12 levels. In this issue of Cell, Shen and colleagues uncover the metabolic role of CLYBL, linking its function to B12 metabolism and the immunomodulatory metabolite, itaconate.


Asunto(s)
Succinatos , Vitamina B 12 , Técnicas de Inactivación de Genes , Humanos , Vitaminas
2.
Cell ; 149(3): 656-70, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22541435

RESUMEN

Tumor maintenance relies on continued activity of driver oncogenes, although their rate-limiting role is highly context dependent. Oncogenic Kras mutation is the signature event in pancreatic ductal adenocarcinoma (PDAC), serving a critical role in tumor initiation. Here, an inducible Kras(G12D)-driven PDAC mouse model establishes that advanced PDAC remains strictly dependent on Kras(G12D) expression. Transcriptome and metabolomic analyses indicate that Kras(G12D) serves a vital role in controlling tumor metabolism through stimulation of glucose uptake and channeling of glucose intermediates into the hexosamine biosynthesis and pentose phosphate pathways (PPP). These studies also reveal that oncogenic Kras promotes ribose biogenesis. Unlike canonical models, we demonstrate that Kras(G12D) drives glycolysis intermediates into the nonoxidative PPP, thereby decoupling ribose biogenesis from NADP/NADPH-mediated redox control. Together, this work provides in vivo mechanistic insights into how oncogenic Kras promotes metabolic reprogramming in native tumors and illuminates potential metabolic targets that can be exploited for therapeutic benefit in PDAC.


Asunto(s)
Adenocarcinoma/metabolismo , Modelos Animales de Enfermedad , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Humanos , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Transcripción Genética
3.
Cell ; 144(4): 601-13, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21295337

RESUMEN

Understanding the factors that impede immune responses to persistent viruses is essential in designing therapies for HIV infection. Mice infected with LCMV clone-13 have persistent high-level viremia and a dysfunctional immune response. Interleukin-7, a cytokine that is critical for immune development and homeostasis, was used here to promote immunity toward clone-13, enabling elucidation of the inhibitory pathways underlying impaired antiviral immune response. Mechanistically, IL-7 downregulated a critical repressor of cytokine signaling, Socs3, resulting in amplified cytokine production, increased T cell effector function and numbers, and viral clearance. IL-7 enhanced thymic output to expand the naive T cell pool, including T cells that were not LCMV specific. Additionally, IL-7 promoted production of cytoprotective IL-22 that abrogated liver pathology. The IL-7-mediated effects were dependent on endogenous IL-6. These attributes of IL-7 have profound implications for its use as a therapeutic in the treatment of chronic viral diseases.


Asunto(s)
Interleucina-7/uso terapéutico , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Animales , Antígenos de Diferenciación/metabolismo , Regulación hacia Abajo , Factores de Transcripción Forkhead/metabolismo , Humanos , Interleucina-6/inmunología , Interleucina-7/inmunología , Ratones , Receptor de Muerte Celular Programada 1 , Proteínas Recombinantes/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Linfocitos T/inmunología
4.
EMBO J ; 38(19): e96659, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31454099

RESUMEN

Loss of the histone H3.3-specific chaperone component ATRX or its partner DAXX frequently occurs in human cancers that employ alternative lengthening of telomeres (ALT) for chromosomal end protection, yet the underlying mechanism remains unclear. Here, we report that ATRX/DAXX does not serve as an immediate repressive switch for ALT. Instead, ATRX or DAXX depletion gradually induces telomere DNA replication dysfunction that activates not only homology-directed DNA repair responses but also cell cycle checkpoint control. Mechanistically, we demonstrate that this process is contingent on ATRX/DAXX histone chaperone function, independently of telomere length. Combined ATAC-seq and telomere chromatin immunoprecipitation studies reveal that ATRX loss provokes progressive telomere decondensation that culminates in the inception of persistent telomere replication dysfunction. We further show that endogenous telomerase activity cannot overcome telomere dysfunction induced by ATRX loss, leaving telomere repair-based ALT as the only viable mechanism for telomere maintenance during immortalization. Together, these findings implicate ALT activation as an adaptive response to ATRX/DAXX loss-induced telomere replication dysfunction.


Asunto(s)
Proteínas Co-Represoras/genética , Chaperonas Moleculares/genética , Homeostasis del Telómero , Telómero/metabolismo , Proteína Nuclear Ligada al Cromosoma X/genética , Línea Celular , Reparación del ADN , Eliminación de Gen , Células HEK293 , Humanos , Telomerasa/metabolismo
5.
Nat Immunol ; 11(7): 618-27, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20467422

RESUMEN

CD4(+) regulatory T cells (T(reg) cells) characterized by expression of the transcription factor Foxp3 have a pivotal role in maintaining immunological tolerance. Here we show that mice with T cell-specific deletion of both the Foxo1 and Foxo3 transcription factors (collectively called 'Foxo proteins' here) developed a fatal multifocal inflammatory disorder due in part to T(reg) cell defects. Foxo proteins functioned in a T(reg) cell-intrinsic manner to regulate thymic and transforming growth factor-beta (TGF-beta)-induced Foxp3 expression, in line with the ability of Foxo proteins to bind to Foxp3 locus and control Foxp3 promoter activity. Transcriptome analyses showed that Foxo proteins regulated the expression of additional T(reg) cell-associated genes and were essential for inhibiting the acquisition of effector T cell characteristics by T(reg) cells. Thus, Foxo proteins have crucial roles in specifying the T(reg) cell lineage.


Asunto(s)
Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/metabolismo , Timo/metabolismo , Animales , Antígenos CD4/biosíntesis , Linaje de la Célula , Células Cultivadas , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Inflamación , Ratones , Ratones Noqueados , Ratones Mutantes , Mutación/genética , Unión Proteica , Elementos Reguladores de la Transcripción/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Timo/inmunología , Timo/patología , Factor de Crecimiento Transformador beta/metabolismo
6.
Genes Dev ; 28(13): 1429-44, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24990963

RESUMEN

The three EglN prolyl hydroxylases (EglN1, EglN2, and EglN3) regulate the stability of the HIF transcription factor. We recently showed that loss of EglN2, however, also leads to down-regulation of Cyclin D1 and decreased cell proliferation in a HIF-independent manner. Here we report that EglN2 can hydroxylate FOXO3a on two specific prolyl residues in vitro and in vivo. Hydroxylation of these sites prevents the binding of USP9x deubiquitinase, thereby promoting the proteasomal degradation of FOXO3a. FOXO transcription factors can repress Cyclin D1 transcription. Failure to hydroxylate FOXO3a promotes its accumulation in cells, which in turn suppresses Cyclin D1 expression. These findings provide new insights into post-transcriptional control of FOXO3a and provide a new avenue for pharmacologically altering Cyclin D1 activity.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular , Células Cultivadas , Ciclina D1/genética , Proteína Forkhead Box O3 , Hidroxilación , Células MCF-7 , Ratones , Unión Proteica , Estabilidad Proteica
7.
Nat Immunol ; 9(6): 623-31, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18488031

RESUMEN

Although the essential role of the adaptor protein SLP-65 in pre-B cell differentiation is established, the molecular mechanism underlying its function is poorly understood. In this study, we uncover a link between SLP-65-dependent signaling and the phosphoinositide-3-OH kinase (PI(3)K)-protein kinase B (PKB)-Foxo pathway. We show that the forkhead box transcription factor Foxo3a promotes light chain rearrangement in pre-B cells. Our data suggest that PKB suppresses light chain recombination by phosphorylating Foxo proteins, whereas reconstitution of SLP-65 function counteracts PKB activation and promotes Foxo3a and Foxo1 activity in pre-B cells. Together, these data illuminate a molecular function of SLP-65 and identify a key role for Foxo proteins in the regulation of light chain recombination, receptor editing and B cell selection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Linfocitos B/inmunología , Factores de Transcripción Forkhead/metabolismo , Genes de las Cadenas Ligeras de las Inmunoglobulinas/genética , Proteínas Tirosina Quinasas/metabolismo , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Recombinación Genética
8.
Genes Dev ; 26(13): 1459-72, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22751500

RESUMEN

Multidimensional cancer genome analysis and validation has defined Quaking (QKI), a member of the signal transduction and activation of RNA (STAR) family of RNA-binding proteins, as a novel glioblastoma multiforme (GBM) tumor suppressor. Here, we establish that p53 directly regulates QKI gene expression, and QKI protein associates with and leads to the stabilization of miR-20a; miR-20a, in turn, regulates TGFßR2 and the TGFß signaling network. This pathway circuitry is substantiated by in silico epistasis analysis of its components in the human GBM TCGA (The Cancer Genome Atlas Project) collection and by their gain- and loss-of-function interactions in in vitro and in vivo complementation studies. This p53-QKI-miR-20a-TGFß pathway expands our understanding of the p53 tumor suppression network in cancer and reveals a novel tumor suppression mechanism involving regulation of specific cancer-relevant microRNAs.


Asunto(s)
Línea Celular , Glioblastoma/metabolismo , MicroARNs/metabolismo , Estabilidad del ARN , Proteínas de Unión al ARN/metabolismo , Animales , Glioblastoma/genética , Humanos , Ratones , MicroARNs/genética , Proteínas de Unión al ARN/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
EMBO J ; 32(19): 2589-602, 2013 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-24013118

RESUMEN

Forkhead Box O (FoxO) transcription factors act in adult stem cells to preserve their regenerative potential. Previously, we reported that FoxO maintains the long-term proliferative capacity of neural stem/progenitor cells (NPCs), and that this occurs, in part, through the maintenance of redox homeostasis. Herein, we demonstrate that among the FoxO3-regulated genes in NPCs are a host of enzymes in central carbon metabolism that act to combat reactive oxygen species (ROS) by directing the flow of glucose and glutamine carbon into defined metabolic pathways. Characterization of the metabolic circuit observed upon loss of FoxO3 revealed a drop in glutaminolysis and filling of the tricarboxylic acid (TCA) cycle. Additionally, we found that glucose uptake, glucose metabolism and oxidative pentose phosphate pathway activity were similarly repressed in the absence of FoxO3. Finally, we demonstrate that impaired glucose and glutamine metabolism compromises the proliferative potential of NPCs and that this is exacerbated following FoxO3 loss. Collectively, our findings show that a FoxO3-dependent metabolic programme supports redox balance and the neurogenic potential of NPCs.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Células-Madre Neurales/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Glucosa/metabolismo , Glutamina/metabolismo , Redes y Vías Metabólicas , Ratones , Ratones Transgénicos , NADP/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Vía de Pentosa Fosfato , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
10.
Nature ; 469(7328): 102-6, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21113150

RESUMEN

An ageing world population has fuelled interest in regenerative remedies that may stem declining organ function and maintain fitness. Unanswered is whether elimination of intrinsic instigators driving age-associated degeneration can reverse, as opposed to simply arrest, various afflictions of the aged. Such instigators include progressively damaged genomes. Telomerase-deficient mice have served as a model system to study the adverse cellular and organismal consequences of wide-spread endogenous DNA damage signalling activation in vivo. Telomere loss and uncapping provokes progressive tissue atrophy, stem cell depletion, organ system failure and impaired tissue injury responses. Here, we sought to determine whether entrenched multi-system degeneration in adult mice with severe telomere dysfunction can be halted or possibly reversed by reactivation of endogenous telomerase activity. To this end, we engineered a knock-in allele encoding a 4-hydroxytamoxifen (4-OHT)-inducible telomerase reverse transcriptase-oestrogen receptor (TERT-ER) under transcriptional control of the endogenous TERT promoter. Homozygous TERT-ER mice have short dysfunctional telomeres and sustain increased DNA damage signalling and classical degenerative phenotypes upon successive generational matings and advancing age. Telomerase reactivation in such late generation TERT-ER mice extends telomeres, reduces DNA damage signalling and associated cellular checkpoint responses, allows resumption of proliferation in quiescent cultures, and eliminates degenerative phenotypes across multiple organs including testes, spleens and intestines. Notably, somatic telomerase reactivation reversed neurodegeneration with restoration of proliferating Sox2(+) neural progenitors, Dcx(+) newborn neurons, and Olig2(+) oligodendrocyte populations. Consistent with the integral role of subventricular zone neural progenitors in generation and maintenance of olfactory bulb interneurons, this wave of telomerase-dependent neurogenesis resulted in alleviation of hyposmia and recovery of innate olfactory avoidance responses. Accumulating evidence implicating telomere damage as a driver of age-associated organ decline and disease risk and the marked reversal of systemic degenerative phenotypes in adult mice observed here support the development of regenerative strategies designed to restore telomere integrity.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/patología , Telomerasa/deficiencia , Telomerasa/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/anatomía & histología , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Daño del ADN/efectos de los fármacos , Proteína Doblecortina , Activación Enzimática/efectos de los fármacos , Reactivadores Enzimáticos/farmacología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Vaina de Mielina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/enzimología , Células-Madre Neurales/patología , Tamaño de los Órganos/efectos de los fármacos , Fenotipo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Medicina Regenerativa , Olfato/efectos de los fármacos , Olfato/fisiología , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Telomerasa/genética , Telómero/efectos de los fármacos , Telómero/metabolismo , Telómero/patología
11.
Cell Mol Life Sci ; 71(21): 4221-41, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25037158

RESUMEN

Metabolic activity indicative of cellular demand is emerging as a key player in cell fate decision. Numerous studies have demonstrated that diverse metabolic pathways have a critical role in the control of the proliferation, differentiation and quiescence of stem cells. The identification of neural stem/progenitor cells (NSPCs) and the characterization of their development and fate decision process have provided insight into the regenerative potential of the adult brain. As a result, the potential of NSPCs in cell replacement therapies for neurological diseases is rapidly growing. The aim of this review is to discuss the recent findings on the crosstalk among key regulators of NSPC development and the metabolic regulation crucial for the function and cell fate decisions of NSPCs. Fundamental understanding of the metabolic circuits in NSPCs may help to provide novel approaches for reactivating neurogenesis to treat degenerative brain conditions and cognitive decline.


Asunto(s)
Regulación de la Expresión Génica , Células-Madre Neurales/citología , Animales , Encéfalo/fisiología , Linaje de la Célula , Cognición , Trastornos del Conocimiento/metabolismo , Epigénesis Genética , Humanos , Hipoxia , Inflamación , Lípidos/química , Enfermedades Neurodegenerativas/metabolismo , Neurogénesis/fisiología , Regeneración
12.
Arch Biochem Biophys ; 534(1-2): 55-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22902436

RESUMEN

Neural stem cells (NSCs) persist over the lifespan of mammals to give rise to committed progenitors and their differentiated cells in order to maintain the brain homeostasis. To this end, NSCs must be able to self-renew and otherwise maintain their quiescence. Suppression of aberrant proliferation or undesired differentiation is crucial to preclude either malignant growth or precocious depletion of NSCs. The PI3K-Akt-FoxO signaling pathway plays a central role in the regulation of multiple stem cells including one in the mammalian brain. In particular, members of FoxO family transcription factors are highly expressed in these stem cells. As an important downstream effector of growth, differentiation, and stress stimuli, mammalian FoxO transcription factor family controls cellular proliferation, oxidative stress response, homeostasis, and eventual maintenance of long-term repopulating potential. The review will focus on the current understanding of FoxO function in NSCs as well as discuss their biological activities that contribute to determining neural stem cell fate.


Asunto(s)
Encéfalo/metabolismo , Factores de Transcripción Forkhead/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Animales , Apoptosis , Autofagia , Encéfalo/citología , Ciclo Celular , Proliferación Celular , Factores de Transcripción Forkhead/genética , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Células-Madre Neurales/citología , Estrés Oxidativo , Mapeo de Interacción de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Vía de Señalización Wnt
13.
Liver Int ; 33(2): 231-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23295054

RESUMEN

BACKGROUND/AIMS: Impaired regulation of apoptosis has been suggested to play a role in the pathogenesis of Primary Biliary Cirrhosis (PBC). In this study, we analysed a signalling pathway that comprises the transcription factor FoxO3a and its downstream target Bim, a Bcl-2 interacting mediator of apoptosis. MATERIALS & METHODS: The tissues examined included livers explanted from patients with cirrhotic PBC, primary sclerosing cholangitis (PSC), alcoholic liver disease (ALD) and liver biopsies from patients with non-cirrhotic PBC. Large margin resections of hepatocellular carcinoma were used as controls. RESULTS: Expression of FoxO3a and Bim mRNA was significantly enhanced in both non-cirrhotic and end-stage PBC (2.2-fold and 4.3-fold increases, respectively), but not in the other disorders. Similarly, FOXO3a protein level was increased in end-stage PBC (P < 0.05 vs. control). A significant increase in Bim mRNA in non-cirrhotic and cirrhotic PBC was observed (2.2-fold and 8.2-fold respectively). In addition, the most pro-apoptotic isoform of Bim dominated in livers of PBC patients (2.5- fold increase vs. control; P < 0.05). Enhanced FoxO3a and Bim expression was associated with a substantial activation of caspase-3 in PBC (2-fold increase vs. controls; P < 0.0001), whereas it was decreased in both ALD and PSC (46% and 67% reductions respectively). The relationship between FoxO3a and Bim was further investigated in the livers of FoxO-deficient mice. The somatic deletion of FoxO3a caused a significant decrease in Bim, but not caspase-3 protein expression confirming the crucial role of FoxO3a in induction of Bim gene transcription. CONCLUSIONS: Our results imply that the FoxO3/Bim signalling pathway can be of importance in the livers of patients with PBC.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/fisiología , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/fisiología , Cirrosis Hepática Biliar/fisiopatología , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Proteína 11 Similar a Bcl2 , Caspasa 3/metabolismo , Colangitis Esclerosante/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Humanos , Cirrosis Hepática Biliar/metabolismo , Hepatopatías Alcohólicas/metabolismo , Ratones , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Proc Natl Acad Sci U S A ; 107(15): 6912-7, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20351267

RESUMEN

Glioblastoma multiforme (GBM) is the most common and lethal primary brain cancer that is driven by aberrant signaling of growth factor receptors, particularly the epidermal growth factor receptor (EGFR). EGFR signaling is tightly regulated by receptor endocytosis and lysosome-mediated degradation, although the molecular mechanisms governing such regulation, particularly in the context of cancer, remain poorly delineated. Here, high-resolution genomic profiles of GBM identified a highly recurrent focal 1p36 deletion encompassing the putative tumor suppressor gene, Mig-6. We show that Mig-6 quells the malignant potential of GBM cells and dampens EGFR signaling by driving EGFR into late endosomes and lysosome-mediated degradation upon ligand stimulation. Mechanistically, this effect is mediated by the binding of Mig-6 to a SNARE protein STX8, a protein known to be required for late endosome trafficking. Thus, Mig-6 functions to ensure recruitment of internalized receptor to late endosomes and subsequently the lysosomal degradation compartment through its ability to specifically link EGFR and STX8 during ligand-stimulated EGFR trafficking. In GBM, the highly frequent loss of Mig-6 would therefore serve to sustain aberrant EGFR-mediated oncogenic signaling. Together, these data uncover a unique tumor suppression mechanism involving the regulation of receptor trafficking.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Neoplasias Encefálicas/metabolismo , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Proteínas Supresoras de Tumor/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lisosomas/metabolismo , Ratones , Invasividad Neoplásica , Proteínas Supresoras de Tumor/genética , Técnicas del Sistema de Dos Híbridos
15.
Dev Dyn ; 241(4): 803-13, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22411556

RESUMEN

BACKGROUND: The forkhead transcription factor FoxO1 is involved in cell cycle regulation during cardiovascular development. Systemic loss of FoxO1 results in lethality at embryonic day 10.5 with severe cardiovascular defects; however, the cell-type-specific requirements for FoxO1 in cardiovascular development are unknown. Here we examine the role of FoxO1 using a conditional loss of function approach. RESULTS: Loss of FoxO1 in differentiated cardiac myocytes has no apparent effect on cardiovascular development. In contrast, endothelial-specific FoxO1 deficiency in Tie2Cre;FoxO1(fl/fl) embryos results in lethality at E10.5, which recapitulates the FoxO1-null phenotype. Tie2Cre;FoxO1(fl/fl) embryos have an intact differentiated endothelium, but display defective remodeling of vasculature. Additional effects on heart development include reduced myocardial trabeculation, which is likely secondary to the endothelial abnormalities, and hypoplasia of endocardial cushions. CONCLUSIONS: The phenotype of Tie2Cre;FoxO1(fl/fl) mutant embryos demonstrates that FoxO1 is required specifically in endothelial cells to regulate formation of the heart and vasculature during development.


Asunto(s)
Linaje de la Célula/genética , Endotelio Vascular/embriología , Factores de Transcripción Forkhead/fisiología , Corazón/embriología , Animales , Diferenciación Celular , Endotelio Vascular/fisiología , Proteína Forkhead Box O1 , Técnicas de Silenciamiento del Gen , Corazón/fisiología , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Especificidad de Órganos
16.
Aging Cell ; 22(3): e13763, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36617632

RESUMEN

Intronic single-nucleotide polymorphisms (SNPs) in FOXO3A are associated with human longevity. Currently, it is unclear how these SNPs alter FOXO3A functionality and human physiology, thereby influencing lifespan. Here, we identify a primate-specific FOXO3A transcriptional isoform, FOXO3A-Short (FOXO3A-S), encoding a major longevity-associated SNP, rs9400239 (C or T), within its 5' untranslated region. The FOXO3A-S mRNA is highly expressed in the skeletal muscle and has very limited expression in other tissues. We find that the rs9400239 variant influences the stability and functionality of the primarily nuclear protein(s) encoded by the FOXO3A-S mRNA. Assessment of the relationship between the FOXO3A-S polymorphism and peripheral glucose clearance during insulin infusion (Rd clamp) in a cohort of Danish twins revealed that longevity T-allele carriers have markedly faster peripheral glucose clearance rates than normal lifespan C-allele carriers. In vitro experiments in human myotube cultures utilizing overexpression of each allele showed that the C-allele represses glycolysis independently of PI3K signaling, while overexpression of the T-allele represses glycolysis only in a PI3K-inactive background. Supporting this finding inducible knockdown of the FOXO3A-S C-allele in cultured myotubes increases the glycolytic rate. We conclude that the rs9400239 polymorphism acts as a molecular switch which changes the identity of the FOXO3A-S-derived protein(s), which in turn alters the relationship between FOXO3A-S and insulin/PI3K signaling and glycolytic flux in the skeletal muscle. This critical difference endows carriers of the FOXO3A-S T-allele with consistently higher insulin-stimulated peripheral glucose clearance rates, which may contribute to their longer and healthier lifespans.


Asunto(s)
Glucosa , Longevidad , Animales , Humanos , Proteína Forkhead Box O3/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Insulina/genética , Insulina/metabolismo , Longevidad/genética , Fosfatidilinositol 3-Quinasas/genética , ARN Mensajero
17.
bioRxiv ; 2023 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-36798426

RESUMEN

Telomere length maintenance is essential for cellular immortalization and tumorigenesis. 5% - 10% of human cancers rely on a recombination-based mechanism termed alternative lengthening of telomeres (ALT) to sustain their replicative immortality, yet there are currently no targeted therapies. Through CRISPR/Cas9-based genetic screens in an ALT-immortalized isogenic cellular model, here we identify histone lysine demethylase KDM2A as a molecular vulnerability selectively for cells contingent on ALT-dependent telomere maintenance. Mechanistically, we demonstrate that KDM2A is required for dissolution of the ALT-specific telomere clusters following homology-directed telomere DNA synthesis. We show that KDM2A promotes de-clustering of ALT multitelomeres through facilitating isopeptidase SENP6-mediated SUMO deconjugation at telomeres. Inactivation of KDM2A or SENP6 impairs post-recombination telomere de-SUMOylation and thus dissolution of ALT telomere clusters, leading to gross chromosome missegregation and mitotic cell death. These findings together establish KDM2A as a selective molecular vulnerability and a promising drug target for ALT-dependent cancers.

18.
Nat Commun ; 14(1): 1756, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36991019

RESUMEN

Telomere length maintenance is essential for cellular immortalization and tumorigenesis. 5% - 10% of human cancers rely on a recombination-based mechanism termed alternative lengthening of telomeres (ALT) to sustain their replicative immortality, yet there are currently no targeted therapies. Through CRISPR/Cas9-based genetic screens in an ALT-immortalized isogenic cellular model, here we identify histone lysine demethylase KDM2A as a molecular vulnerability selectively for cells contingent on ALT-dependent telomere maintenance. Mechanistically, we demonstrate that KDM2A is required for dissolution of the ALT-specific telomere clusters following recombination-directed telomere DNA synthesis. We show that KDM2A promotes de-clustering of ALT multitelomeres through facilitating isopeptidase SENP6-mediated SUMO deconjugation at telomeres. Inactivation of KDM2A or SENP6 impairs post-recombination telomere de-SUMOylation and thus dissolution of ALT telomere clusters, leading to gross chromosome missegregation and mitotic cell death. These findings together establish KDM2A as a selective molecular vulnerability and a promising drug target for ALT-dependent cancers.


Asunto(s)
Proteínas F-Box , Neoplasias , Telomerasa , Humanos , Línea Celular , ADN , Homeostasis del Telómero/genética , Telómero/genética , Telómero/metabolismo , Neoplasias/genética , Telomerasa/genética , Cisteína Endopeptidasas/metabolismo , Proteínas F-Box/genética , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo
19.
Blood Adv ; 7(20): 6211-6224, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37327122

RESUMEN

Mantle cell lymphoma (MCL) is an incurable B-cell malignancy that comprises up to 6% of non-Hodgkin lymphomas diagnosed annually and is associated with a poor prognosis. The average overall survival of patients with MCL is 5 years, and for most patients who progress on targeted agents, survival remains at a dismal 3 to 8 months. There is a major unmet need to identify new therapeutic approaches that are well tolerated to improve treatment outcomes and quality of life. The protein arginine methyltransferase 5 (PRMT5) enzyme is overexpressed in MCL and promotes growth and survival. Inhibition of PRMT5 drives antitumor activity in MCL cell lines and preclinical murine models. PRMT5 inhibition reduced the activity of prosurvival AKT signaling, which led to the nuclear translocation of FOXO1 and modulation of its transcriptional activity. Chromatin immunoprecipitation and sequencing identified multiple proapoptotic BCL-2 family members as FOXO1-bound genomic loci. We identified BAX as a direct transcriptional target of FOXO1 and demonstrated its critical role in the synergy observed between the selective PRMT5 inhibitor, PRT382, and the BCL-2 inhibitor, venetoclax. Single-agent and combination treatments were performed in 9 MCL lines. Loewe synergy scores showed significant levels of synergy in most MCL lines tested. Preclinical, in vivo evaluation of this strategy in multiple MCL models showed therapeutic synergy with combination venetoclax/PRT382 treatment with an increased survival advantage in 2 patient-derived xenograft models (P ≤ .0001, P ≤ .0001). Our results provide mechanistic rationale for the combination of PRMT5 inhibition and venetoclax to treat patients with MCL.


Asunto(s)
Antineoplásicos , Compuestos Bicíclicos Heterocíclicos con Puentes , Linfoma de Células del Manto , Sulfonamidas , Animales , Humanos , Ratones , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Linfoma de Células del Manto/tratamiento farmacológico , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Calidad de Vida
20.
J Biol Chem ; 286(9): 7468-78, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21159781

RESUMEN

Transcriptional regulatory mechanisms of cardiac oxidative stress resistance are not well defined. FoxO transcription factors are critical mediators of oxidative stress resistance in multiple cell types, but cardioprotective functions have not been reported previously. FoxO function in oxidative stress resistance was investigated in cultured cardiomyocytes and in mice with cardiomyocyte-specific combined deficiency of FoxO1 and FoxO3 subjected to myocardial infarction (MI) or acute ischemia/reperfusion (I/R) injury. Induction of oxidative stress in cardiomyocytes promotes FoxO1 and FoxO3 nuclear localization and target gene activation. Infection of cardiomyocytes with a dominant-negative FoxO1(Δ256) adenovirus results in a significant increase in reactive oxygen species and cell death, whereas increased FoxO1 or FoxO3 expression reduces reactive oxygen species and cell death. Mice generated with combined conditional deletion of FoxO1 and FoxO3 specifically in cardiomyocytes were subjected to I/R or MI. Loss of FoxO1 and FoxO3 in cardiomyocytes results in a significant increase in infarct area with decreased expression of the antiapoptotic molecules, PTEN-induced kinase1 (PINK1) and CBP/P300-interacting transactivator (CITED2). Expressions of the antioxidants catalase and manganese superoxide dismutase-2 (SOD2) and the autophagy-related proteins LC3II and Gabarapl1 also are decreased following I/R compared with controls. Mice with cardiomyocyte-specific FoxO deficiency subjected to MI have reduced cardiac function, increased scar formation, induction of stress-responsive signaling, and increased apoptotic cell death relative to controls. These data support a critical role for FoxOs in promoting cardiomyocyte survival during conditions of oxidative stress through induction of antioxidants and cell survival pathways.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/fisiología , Animales , Animales Recién Nacidos , Antioxidantes/metabolismo , Apoptosis/fisiología , Autofagia/fisiología , Proteínas de Ciclo Celular/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Mutantes , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/citología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Oxígeno/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA