Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
Cell ; 139(3): 573-86, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19879843

RESUMEN

Previous work has shown that mature B cells depend upon survival signals delivered to the cells by their antigen receptor (BCR). To identify the molecular nature of this survival signal, we have developed a genetic approach in which ablation of the BCR is combined with the activation of specific, BCR dependent signaling cascades in mature B cells in vivo. Using this system, we provide evidence that the survival of BCR deficient mature B cells can be rescued by a single signaling pathway downstream of the BCR, namely PI3K signaling, with the FOXO1 transcription factor playing a central role.


Asunto(s)
Linfocitos B/citología , Linfocitos B/metabolismo , Supervivencia Celular , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Ratones , Ratones Noqueados , Transducción de Señal
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Angiogenesis ; 16(4): 759-72, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23677673

RESUMEN

Peripheral artery disease (PAD) is characterized by chronic muscle ischemia. Compensatory angiogenesis is minimal within ischemic muscle despite an increase in angiogenic factors. This may occur due to the prevalence of angiostatic factors. Regulatory mechanisms that could evoke an angiostatic environment during ischemia are largely unknown. Forkhead box O (FoxO) transcription factors, known to repress endothelial cell proliferation in vitro, are potential candidates. Our goal was to determine whether FoxO proteins promote an angiostatic phenotype within ischemic muscle. FoxO1 and the angiostatic matrix protein thrombospondin 1 (THBS1) were elevated in ischemic muscle from PAD patients, or from mice post-femoral artery ligation. Mice with conditional endothelial cell-directed deletion of FoxO proteins (Mx1Cre (+), FoxO1,3,4 (L/L) , referred to as FoxOΔ) were used to assess the role of endothelial FoxO proteins within ischemic tissue. FoxO deletion abrogated the elevation of FoxO1 and THBS1 proteins, enhanced hindlimb blood flow recovery and improved neovascularization in murine ischemic muscle. Endothelial cell outgrowth from 3D explant cultures was more robust in muscles derived from FoxOΔ mice. FoxO1 overexpression induced THBS1 production, and a direct interaction of endogenous FoxO1 with the THBS1 promoter was detectable in primary endothelial cells. We provide evidence that FoxO1 directly regulates THBS1 within ischemic muscle. Altogether, these findings bring novel insight into the regulatory mechanisms underlying the repression of angiogenesis within peripheral ischemic tissues.


Asunto(s)
Endotelio Vascular/metabolismo , Factores de Transcripción Forkhead/fisiología , Isquemia/fisiopatología , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Enfermedad Arterial Periférica/metabolismo , Trombospondina 1/biosíntesis , Anciano , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Arteria Femoral , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/deficiencia , Eliminación de Gen , Regulación de la Expresión Génica , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/etiología , Isquemia/genética , Ligadura , Ratones , Persona de Mediana Edad , Enfermedad Arterial Periférica/complicaciones , Enfermedad Arterial Periférica/fisiopatología , Factores de Riesgo , Organismos Libres de Patógenos Específicos , Trombospondina 1/genética , Regulación hacia Arriba
10.
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
11.
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
12.
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
13.
Proc Natl Acad Sci U S A ; 105(49): 19372-7, 2008 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-19050074

RESUMEN

Pancreas ductal adenocarcinoma (PDAC) is a highly lethal cancer that typically presents as advanced, unresectable disease. This invasive tendency, coupled with intrinsic resistance to standard therapies and genome instability, are major contributors to poor long-term survival. The genetic elements governing the invasive propensity of PDAC have not been well elucidated. Here, in the course of validating resident genes in highly recurrent and focal amplifications in PDAC, we have identified Rio Kinase 3 (RIOK3) as an amplified gene that alters cytoskeletal architecture as well as promotes pancreatic ductal cell migration and invasion. We determined that RIOK3 promotes its invasive activities through activation of the small G protein, Rac. This genomic and functional link to Rac signaling prompted a genome wide survey of other components of the Rho family network, revealing p21 Activated Kinase 4 (PAK4) as another amplified gene in PDAC tumors and cell lines. Like RIOK3, PAK4 promotes pancreas ductal cell motility and invasion. Together, the genomic and functional profiles establish the Rho family GTP-binding proteins as integral to the hallmark invasive nature of this lethal disease.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Conductos Pancreáticos/fisiología , Neoplasias Pancreáticas/genética , Proteínas Serina-Treonina Quinasas/genética , Quinasas p21 Activadas/genética , Proteínas de Unión al GTP rho/genética , Animales , Carcinoma Ductal Pancreático/patología , Línea Celular Transformada , Movimiento Celular/fisiología , Regulación Neoplásica de la Expresión Génica , Genómica , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Conductos Pancreáticos/citología , Neoplasias Pancreáticas/patología , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rho/metabolismo
14.
J Microbiol Biotechnol ; 20(9): 1295-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20890094

RESUMEN

Recently, recombinant Streptomyces venezuelae has been established as a heterologous host for microbial production of flavanones and stilbenes, a class of plant-specific polyketides. In the present work, we expanded the applicability of the S. venezuelae system to the production of more diverse plant polyketides including flavones and flavonols. A plasmid with the synthetic codon-optimized flavone synthase I gene from Petroselium crispum was introduced to S. venezuelae DHS2001 bearing a deletion of the native pikromycin polyketide synthase gene, and the resulting strain generated flavones from exogenously fed flavanones. In addition, a recombinant S. venezuelae mutant expressing a codon-optimized flavanone 3beta-hydroxylase gene from Citrus siensis and a flavonol synthase gene from Citrus unshius also successfully produced flavonols.


Asunto(s)
Flavonas/biosíntesis , Flavonoles/biosíntesis , Microbiología Industrial/métodos , Streptomyces/metabolismo , Apiaceae/enzimología , Apiaceae/genética , Citrus/enzimología , Expresión Génica , Genes de Plantas/genética , Ingeniería Genética , Oxigenasas de Función Mixta/genética , Streptomyces/genética
15.
Cell Metab ; 32(5): 889-900.e7, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33147486

RESUMEN

Differential WNT and Notch signaling regulates differentiation of Lgr5+ crypt-based columnar cells (CBCs) into intestinal cell lineages. Recently we showed that mitochondrial activity supports CBCs, while adjacent Paneth cells (PCs) show reduced mitochondrial activity. This implies that CBC differentiation into PCs involves a metabolic transition toward downregulation of mitochondrial dependency. Here we show that Forkhead box O (FoxO) transcription factors and Notch signaling interact in determining CBC fate. In agreement with the organoid data, Foxo1/3/4 deletion in mouse intestine induces secretory cell differentiation. Importantly, we show that FOXO and Notch signaling converge on regulation of mitochondrial fission, which in turn provokes stem cell differentiation into goblet cells and PCs. Finally, scRNA-seq-based reconstruction of CBC differentiation trajectories supports the role of FOXO, Notch, and mitochondria in secretory differentiation. Together, this points at a new signaling-metabolic axis in CBC differentiation and highlights the importance of mitochondria in determining stem cell fate.


Asunto(s)
Células Caliciformes , Intestinos/citología , Mitocondrias/metabolismo , Células de Paneth , Células Madre , Animales , Diferenciación Celular , Línea Celular , Factores de Transcripción Forkhead/metabolismo , Células Caliciformes/citología , Células Caliciformes/metabolismo , Ratones , Dinámicas Mitocondriales , Células de Paneth/citología , Células de Paneth/metabolismo , Receptores Notch/metabolismo , Células Madre/citología , Células Madre/metabolismo
16.
iScience ; 23(4): 101006, 2020 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-32268281

RESUMEN

Design of tissue-specific contrast agents to delineate tumors from background tissues is a major unmet clinical need for ultimate surgical interventions. Bioconjugation of fluorophore(s) to a ligand has been mainly used to target overexpressed receptors on tumors. However, the size of the final targeted ligand can be large, >20 kDa, and cannot readily cross the microvasculature to meet the specific tissue, resulting in low targetability with a high background. Here, we report a small and hydrophilic phenoxazine with high targetability and retention to pancreatic neuroendocrine tumor. This bioengineered fluorophore permits sensitive detection of ultrasmall (<0.5 mm) ectopic tumors within a few seconds after a single bolus injection, highlighting every tumor in the pancreas from the surrounding healthy tissues with reasonable half-life. The knowledge-based approach and validation used to develop structure-inherent tumor-targeted fluorophores have a tremendous potential to improve treatment outcome by providing definite tumor margins for image-guided surgery.

17.
Mol Cell Biol ; 26(19): 7211-23, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16980623

RESUMEN

Sphingosine kinase (Sphk) enzymes are important in intracellular sphingolipid metabolism as well as in the biosynthesis of sphingosine 1-phosphate (S1P), an extracellular lipid mediator. Here, we show that Sphk1 is expressed and is required for small intestinal tumor cell proliferation in Apc Min/+ mice. Adenoma size but not incidence was dramatically reduced in Apc Min/+ Sphk(-/-) mice. Concomitantly, epithelial cell proliferation in the polyps was significantly attenuated, suggesting that Sphk1 regulates adenoma progression. Although the S1P receptors (S1P1R, S1P2R, and S1P3R) are expressed, polyp incidence or size was unaltered in Apc Min/+ S1p2r(-/-), Apc Min/+ S1p3r(-/-), and Apc Min/+ S1p1r(+/-) bigenic mice. These data suggest that extracellular S1P signaling via its receptors is not involved in adenoma cell proliferation. Interestingly, tissue sphingosine content was elevated in the adenomas of Apc Min/+ Sphk1(-/-) mice, whereas S1P levels were not significantly altered. Concomitantly, epithelial cell proliferation and the expression of the G1/S cell cycle regulator CDK4 and c-myc were diminished in the polyps of Apc Min/+ Sphk1(-/-) mice. In rat intestinal epithelial (RIE) cells in vitro, Sphk1 overexpression enhanced cell cycle traverse at the G1/S boundary. In addition, RIE cells treated with sphingosine but not C6-ceramide exhibited reduced cell proliferation, reduced retinoblastoma protein phosphorylation, and cyclin-dependent kinase 4 (Cdk4) expression. Our findings suggest that Sphk1 plays a critical role in intestinal tumor cell proliferation and that inhibitors of Sphk1 may be useful in the control of intestinal cancer.


Asunto(s)
Pólipos Adenomatosos/enzimología , Pólipos Adenomatosos/patología , Neoplasias Gastrointestinales/enzimología , Neoplasias Gastrointestinales/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Pólipos Adenomatosos/genética , Animales , Apoptosis/fisiología , Proteínas de Ciclo Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular , Regulación hacia Abajo/genética , Fase G1/fisiología , Neoplasias Gastrointestinales/genética , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/patología , Lisofosfolípidos/metabolismo , Ratones , Neovascularización Patológica , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de Lisoesfingolípidos/genética , Fase S/fisiología , Esfingosina/análogos & derivados , Esfingosina/metabolismo
18.
Metabolism ; 91: 43-52, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30500562

RESUMEN

PURPOSE: While leptin has been associated with various psycho-physiological functions, the molecular network in leptin-mediated mood regulation remains elusive. METHODS: Anxiolytic behaviors and tyrosine hydroxylase (TH) levels were examined after leptin administration. Functional roles of STAT3 and FoxO1 in regulation of TH expression were investigated using in vivo and in vitro systems. A series of animal behavioral tests using dopaminergic neuron-specific FoxO1 KO (FoxO1 KODAT) were performed and investigated the roles of FoxO1 in regulation of mood behaviors. RESULTS: Here, we show that administration of leptin induces anxiolytic-like phenotype through the activation of signal transducer and activator of transcription 3 (STAT3) and the inhibition of forkhead box protein O1 (FoxO1) in dopaminergic (DA) neurons of the midbrain. Specifically, STAT3 and FoxO1 directly bind to and exert opposing effects on tyrosine hydroxylase (TH) expression, where STAT3 acts as an enhancer and FoxO1 acts as a prominent repressor. Accordingly, suppression of the prominent suppressor FoxO1 by leptin strongly increased TH expression. Furthermore, our previous results showed that specific deletion of FoxO1 in DA neurons (FoxO1 KODAT) led to a profound elevation of TH activity and dopamine contents. Finally, FoxO1 KODAT mice exhibited enhanced leptin sensitivity as well as displayed reduced anxiety- and depression-like behaviors. CONCLUSIONS: This work establishes a novel molecular mechanism of mood behavior regulation by leptin and suggests FoxO1 suppression by leptin might be a key for leptin-induced behavioral manifestation in DA neurons.


Asunto(s)
Afecto/efectos de los fármacos , Proteína Forkhead Box O1/antagonistas & inhibidores , Proteína Forkhead Box O1/metabolismo , Leptina/farmacología , Tirosina 3-Monooxigenasa/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo , Animales , Ansiedad/genética , Ansiedad/psicología , Depresión/metabolismo , Depresión/psicología , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Masculino , Mesencéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Factor de Transcripción STAT3/metabolismo
19.
J Clin Invest ; 114(8): 1082-9, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15489955

RESUMEN

Angiogenesis, or new blood vessel formation, is critical for the growth and spread of tumors. Multiple phases of this process, namely, migration, proliferation, morphogenesis, and vascular stabilization, are needed for optimal tumor growth beyond a diffusion-limited size. The sphingosine 1-phosphate (S1P) receptor-1 (S1P(1)) is required for stabilization of nascent blood vessels during embryonic development. Here we show that S1P(1) expression is strongly induced in tumor vessels. We developed a multiplex RNA interference technique to downregulate S1P(1) in mice. The small interfering RNA (siRNA) for S1P(1) specifically silenced the cognate transcript in endothelial cells and inhibited endothelial cell migration in vitro and the growth of neovessels into subcutaneous implants of Matrigel in vivo. Local injection of S1P(1) siRNA, but not a negative control siRNA, into established tumors inhibited the expression of S1P(1) polypeptide on neovessels while concomitantly suppressing vascular stabilization and angiogenesis, which resulted in dramatic suppression of tumor growth in vivo. These data suggest that S1P(1) is a critical component of the tumor angiogenic response and argue for the utility of siRNA technology in antiangiogenic therapeutics.


Asunto(s)
Neoplasias/irrigación sanguínea , Neovascularización Patológica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Células Cultivadas , Colágeno/metabolismo , Combinación de Medicamentos , Células Endoteliales/citología , Células Endoteliales/fisiología , Regulación Neoplásica de la Expresión Génica , Laminina/metabolismo , Ratones , Trasplante de Neoplasias , Neoplasias/metabolismo , Proteoglicanos/metabolismo , ARN Interferente Pequeño/genética , Receptores de Lisoesfingolípidos/genética , Trasplante Heterólogo
20.
Cancer Res ; 65(9): 3788-95, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867375

RESUMEN

Sphingosine 1-phosphate (S1P) is a lysophospholipid that exerts a variety of responses in cells such as proliferation, migration, and survival. These effects are mediated by G protein-coupled receptors on the cell surface (S1P1-5), which activate downstream signaling intermediates such as Rac and Rho GTPases. Mechanisms of S1P action in human glioblastoma cells are not well defined. S1P receptors (1-5) and S1P-metabolizing enzymes were expressed in three human glioblastoma cell lines. S1P had a profound and differential effect on glioblastoma cell migration. U87 cells treated with S1P showed a significant increase in migration, whereas U118 and U138 cell lines were strongly inhibited. S1P-mediated inhibition correlated with S1P2 receptor expression. FTY720-P, an S1P analogue that binds all S1P receptors except S1P2, did not inhibit glioblastoma cell migration. Overexpression of S1P2 further suppressed migration, and blockage of S1P2 mRNA expression by small interfering RNA reversed the inhibitory effect. Contrary to previous reports showing bimodal regulation of Rac activity and migration by S1P2 receptor stimulation, both Rac1 and RhoA GTPases were activated by S1P treatment in native cells and cells overexpressing S1P2. Treatment of U118 cells with the Rho-associated protein kinase (ROCK) inhibitor Y-27632 restored migration suggesting that ROCK-dependent mechanisms are important. Actin staining of S1P stimulated U118 cells overexpressing beta-galactosidase resulted in pronounced stress fiber formation that was exacerbated by S1P2 overexpression, partially blocked by S1P1, or totally abolished by pretreatment with Y-27632. These data provide evidence of a novel mechanism of S1P inhibition of tumor cell migration via Rho kinase-dependent pathway.


Asunto(s)
Movimiento Celular/fisiología , Glioblastoma/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Lisoesfingolípidos/fisiología , Esfingolípidos/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Amidas/farmacología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Glioblastoma/enzimología , Glioblastoma/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/biosíntesis , Receptores de Lisoesfingolípidos/genética , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/metabolismo , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA