Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
mBio ; 8(5)2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-29018115

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with the human malignancy Kaposi's sarcoma and the lymphoproliferative disorders primary effusion lymphoma and multicentric Castleman's disease. KSHV establishes lytic infection of monocytes in vivo, which may represent an important cellular reservoir during KS disease progression. KS tumors consist of latently infected endothelial cells; however, lytic phase gene products are important for KS onset. Early KS lesion progression is driven by proinflammatory cytokines supplied by immune cell infiltrates including T cells and monocytes. KSHV-infected monocytes may supply the lytic viral products and the inflammatory milieu conducive to KS tumor progression. To establish successful infection, KSHV extensively modulates the host immune system. KSHV antigens activate both innate and adaptive immune responses including KSHV-specific T cells, but lifelong infection is still established. Programmed death ligand 1 (PD-L1) is a prosurvival cell surface protein that suppresses T-cell-mediated killing. PD-L1 is variably present on various tumor cells and is a targetable marker for cancer treatment. We show that KSHV infection of human monocytes increases PD-L1 expression and transcription in a dose-dependent manner. We also saw evidence of lytic gene expression in the KSHV-infected monocytes. Intact KSHV is needed for full PD-L1 response in human monocytes. KSHV induces a general proinflammatory cytokine milieu including interleukins 1α, 1ß, and 6, which have been implicated in early KS lesion progression. KSHV-mediated PD-L1 increase may represent a novel mechanism of KSHV-mediated immune modulation to allow for virus survival and eventually malignant progression.IMPORTANCE KSHV is the etiologic agent of Kaposi's sarcoma and the lymphoproliferative disorders primary effusion lymphoma and multicentric Castleman's disease. Programmed death ligand 1 (PD-L1) is an immunosuppressive cell surface marker that inhibits T cell activation. We report that KSHV infection of primary human monocytes upregulates PD-L1 transcription and protein expression. Analysis of the cytokine and chemokine milieu following KSHV infection of monocytes revealed that KSHV induces interleukins 1α, 1ß, and 6, all of which have been implicated in KS development. Our work has identified another potential immune evasion strategy for KSHV and a potential target for immunotherapy of KSHV-derived disease.


Asunto(s)
Antígeno B7-H1/genética , Citocinas/genética , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Monocitos/inmunología , Monocitos/virología , Citocinas/inmunología , Regulación Viral de la Expresión Génica , Humanos , Inmunidad Innata , Regulación hacia Arriba , Proteínas Virales/genética , Proteínas Virales/inmunología , Latencia del Virus
2.
J Virol ; 89(22): 11572-83, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26355087

RESUMEN

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) is a gammaherpesvirus known to establish lifelong latency in the human host. We and others have previously shown that three KSHV homologs of cellular interferon regulatory factors (IRFs), known as viral IRFs (vIRFs), participate in evasion of the host interferon (IFN) response. We report that vIRF1 interacts with the cellular interferon-stimulated gene 15 (ISG15) E3 ligase, HERC5, in the context of Toll-like receptor 3 (TLR3) activation and IFN induction. The ISG15 protein is covalently conjugated to target proteins upon activation of the interferon response. Interaction between vIRF1 and HERC5 was confirmed by immunoprecipitation, and the region between amino acids 224 and 349 of vIRF1 was required for interaction with HERC5. We further report that expression of vIRF1 in the context of TLR3 activation results in decreased ISG15 conjugation of proteins. Specifically, TLR3-induced ISG15 conjugation and protein levels of cellular IRF3, a known ISG15 target, were decreased in the presence of vIRF1 compared to the control. vIRF1 itself was also identified as a target of ISG15 conjugation. KSHV-infected cells exhibited increased ISG15 conjugation upon reactivation from latency in coordination with increased IFN. Furthermore, knockdown of ISG15 in latently infected cells resulted in a higher level of KSHV reactivation and an increase in infectious virus. These data suggest that the KSHV vIRF1 protein affects ISG15 conjugation and interferon responses and may contribute to effective KSHV replication. IMPORTANCE: The KSHV vIRF1 protein can inhibit interferon activation in response to viral infection. We identified a cellular protein named HERC5, which is the major ligase for ISG15, as a vIRF1 binding partner. vIRF1 association with HERC5 altered ISG15 modification of cellular proteins, and knockdown of ISG15 augmented reactivation of KSHV from latency.


Asunto(s)
Citocinas/metabolismo , Herpesvirus Humano 8/fisiología , Factores Reguladores del Interferón/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptor Toll-Like 3/metabolismo , Ubiquitinas/metabolismo , Proteínas Virales/metabolismo , Activación Viral , Secuencia de Aminoácidos , Citocinas/genética , Activación Enzimática , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Interferencia de ARN , ARN Interferente Pequeño , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/genética , Latencia del Virus , Replicación Viral
3.
Proc Natl Acad Sci U S A ; 112(31): E4306-15, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26199418

RESUMEN

Infection of cells with DNA viruses triggers innate immune responses mediated by DNA sensors. cGMP-AMP synthase (cGAS) is a key DNA sensor that produces the cyclic dinucleotide cGMP-AMP (cGAMP) upon activation, which binds to and activates stimulator of interferon genes (STING), leading to IFN production and an antiviral response. Kaposi's sarcoma-associated herpesvirus (KSHV) is a DNA virus that is linked to several human malignancies. We report that KSHV infection activates the cGAS-STING pathway, and that cGAS and STING also play an important role in regulating KSHV reactivation from latency. We screened KSHV proteins for their ability to inhibit this pathway and identified six viral proteins that block IFN-ß activation through this pathway. This study is the first report identifying multiple viral proteins encoded by a human DNA virus that inhibit the cGAS-STING DNA sensing pathway. One such protein, viral interferon regulatory factor 1 (vIRF1), targets STING by preventing it from interacting with TANK binding kinase 1 (TBK1), thereby inhibiting STING's phosphorylation and concomitant activation, resulting in an inhibition of the DNA sensing pathway. Our data provide a unique mechanism for the negative regulation of STING-mediated DNA sensing. Moreover, the depletion of vIRF1 in the context of KSHV infection prevented efficient viral reactivation and replication, and increased the host IFN response to KSHV. The vIRF1-expressing cells also inhibited IFN-ß production following infection with DNA pathogens. Collectively, our results demonstrate that gammaherpesviruses encode inhibitors that block cGAS-STING-mediated antiviral immunity, and that modulation of this pathway is important for viral transmission and the lifelong persistence of herpesviruses in the human population.


Asunto(s)
ADN Viral/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Secuencia de Bases , ADN Viral/genética , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Factor 1 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Datos de Secuencia Molecular , Motivos de Nucleótidos/genética , Sistemas de Lectura Abierta/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Virales/metabolismo , Activación Viral , Latencia del Virus
4.
J Virol ; 88(10): 5778-87, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24623417

RESUMEN

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) has been shown to be recognized by two families of pattern recognition receptors (PRRs), Toll-like receptors (TLRs) and NOD-like receptors (NLRs). Here we show that MAVS and RIG-I (retinoic acid-inducible gene 1), an RLR family member, also have a role in suppressing KSHV replication and production. In the context of primary infection, we show that in cells with depleted levels of MAVS or RIG-I, KSHV transcription is increased, while beta interferon (IFN-ß) induction is attenuated. We also observed that MAVS and RIG-I are critical during the process of reactivation. Depletion of MAVS and RIG-I prior to reactivation led to increased viral load and production of infectious virus. Finally, MAVS depletion in latent KSHV-infected B cells leads to increased viral gene transcription. Overall, this study suggests a role for MAVS and RIG-I signaling during different stages of the KSHV life cycle. IMPORTANCE: We show that RIG-I and its adaptor protein, MAVS, can sense KSHV infection and that these proteins can suppress KSHV replication following primary infection and/or viral reactivation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , ARN Helicasas DEAD-box/metabolismo , Herpesvirus Humano 8/inmunología , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Replicación Viral , Línea Celular , Proteína 58 DEAD Box , Humanos , Receptores Inmunológicos , Transducción de Señal , Activación Viral
5.
J Virol ; 87(2): 798-806, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23115281

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) infection is correlated with three human malignancies and can establish lifelong latent infection in multiple cell types within its human host. In order to establish and maintain infection, KSHV utilizes multiple mechanisms to evade the host immune response. One such mechanism is the expression of a family of genes with homology to cellular interferon (IFN) regulatory factors (IRFs), known as viral IRFs (vIRFs). We demonstrate here that KSHV vIRF1, -2, and -3 have a differential ability to block type I interferon signaling mediated by Toll-like receptor 3 (TLR3), a receptor we have previously shown to be activated upon KSHV infection. vIRF1, -2, and -3 inhibited TLR3-driven activation of IFN transcription reporters. However, only vIRF1 and vIRF2 inhibited increases in both IFN-ß message and protein levels following TLR3 activation. The expression of vIRF1 and vIRF2 also allowed for increased replication of a virus known to activate TLR3 signaling. Furthermore, vIRF1 and vIRF2 may block TLR3-mediated signaling via different mechanisms. Altogether, this report indicates that vIRFs are able to block IFN mediated by TLRs but that each vIRF has a unique function and mechanism for blocking antiviral IFN responses.


Asunto(s)
Herpesvirus Humano 8/patogenicidad , Evasión Inmune , Factores Reguladores del Interferón/metabolismo , Interferones/antagonistas & inhibidores , Receptor Toll-Like 3/antagonistas & inhibidores , Proteínas Virales/metabolismo , Línea Celular , Herpesvirus Humano 8/inmunología , Humanos , Factores Reguladores del Interferón/inmunología , Interferones/inmunología , Receptor Toll-Like 3/inmunología , Proteínas Virales/inmunología
6.
Proc Natl Acad Sci U S A ; 109(29): 11818-23, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22752304

RESUMEN

The metabolic differences between B-NHL and primary human B cells are poorly understood. Among human B-cell non-Hodgkin lymphomas (B-NHL), primary effusion lymphoma (PEL) is a unique subset that is linked to infection with Kaposi's sarcoma-associated herpesvirus (KSHV). We report that the metabolic profiles of primary B cells are significantly different from that of PEL. Compared with primary B cells, both aerobic glycolysis and fatty acid synthesis (FAS) are up-regulated in PEL and other types of nonviral B-NHL. We found that aerobic glycolysis and FAS occur in a PI3K-dependent manner and appear to be interdependent. PEL overexpress the fatty acid synthesizing enzyme, FASN, and both PEL and other B-NHL were much more sensitive to the FAS inhibitor, C75, than primary B cells. Our findings suggest that FASN may be a unique candidate for molecular targeted therapy against PEL and other B-NHL.


Asunto(s)
Linfocitos B/metabolismo , Ácidos Grasos/biosíntesis , Glucólisis/fisiología , Linfoma de Células B/metabolismo , Linfoma de Efusión Primaria/metabolismo , Redes y Vías Metabólicas/fisiología , Transducción de Señal/fisiología , 4-Butirolactona/análogos & derivados , 4-Butirolactona/farmacología , Biología Computacional , Ácido Graso Sintasas/antagonistas & inhibidores , Humanos , Immunoblotting , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
Front Immunol ; 2: 19, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22566809

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is a large double-stranded DNA gammaherpesvirus, and the etiological agent for three human malignancies: Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. To establish and maintain infection, KSHV has evolved unique mechanisms to evade the host immune response. Cellular interferon regulatory factors (IRFs) are a critical part of the host anti-viral immune response. KSHV encodes four homologs of IRFs, vIRF1-4, which inhibit the activity of their cellular counterparts. vIRF1, 2, and 3 have been shown to interact directly with cellular IRFs. Additionally, the vIRFs have other functions such as modulation of Myc, p53, Notch, transforming growth factor-ß, and NF-κB signaling. These activities of vIRFs may contribute to KSHV tumorigenesis. KSHV vIRF1 and vIRF3 have been implicated as oncogenes, making the understanding of KSHV vIRF function vital to understanding KSHV pathogenesis.

8.
J Biol Chem ; 286(7): 5921-33, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21159778

RESUMEN

The PI3K/Akt pathway is activated in stimulated cells and in many cancers to promote glucose metabolism and prevent cell death. Although inhibition of Akt-mediated cell survival may provide a means to eliminate cancer cells, this survival pathway remains incompletely understood. In particular, unlike anti-apoptotic Bcl-2 family proteins that prevent apoptosis independent of glucose, Akt requires glucose metabolism to inhibit cell death. This glucose dependence may occur in part through metabolic regulation of pro-apoptotic Bcl-2 family proteins. Here, we show that activated Akt relies on glycolysis to inhibit induction of Puma, which was uniquely sensitive to metabolic status among pro-apoptotic Bcl-2 family members and was rapidly up-regulated in glucose-deficient conditions. Importantly, preventing Puma expression was critical for Akt-mediated cell survival, as Puma deficiency protected cells from glucose deprivation and Akt could not readily block Puma-mediated apoptosis. In contrast, the pro-apoptotic Bcl-2 family protein Bim was induced normally even when constitutively active Akt was expressed, yet Akt could provide protection from Bim cytotoxicity. Up-regulation of Puma appeared mediated by decreased availability of mitochondrial metabolites rather than glycolysis itself, as alternative mitochondrial fuels could suppress Puma induction and apoptosis upon glucose deprivation. Metabolic regulation of Puma was mediated through combined p53-dependent transcriptional induction and control of Puma protein stability, with Puma degraded in nutrient-replete conditions and long lived in nutrient deficiency. Together, these data identify a key role for Bcl-2 family proteins in Akt-mediated cell survival that may be critical in normal immunity and in cancer through Akt-dependent stimulation of glycolysis to suppress Puma expression.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Apoptosis , Regulación Leucémica de la Expresión Génica , Glucosa/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Supervivencia Celular/genética , Glucosa/genética , Glucólisis/genética , Humanos , Células Jurkat , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba/genética
9.
J Biol Chem ; 283(52): 36344-53, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18990690

RESUMEN

Growth factor stimulation and oncogenic transformation lead to increased glucose metabolism that may provide resistance to cell death. We have previously demonstrated that elevated glucose metabolism characteristic of stimulated or cancerous cells can stabilize the anti-apoptotic Bcl-2 family protein Mcl-1 through inhibition of GSK-3. Here we show that the pro-apoptotic Bcl-2 family protein, Puma, is also metabolically regulated. Growth factor deprivation led to the loss of glucose uptake and induction of Puma. Maintenance of glucose uptake after growth factor withdrawal by expression of the glucose transporter, Glut1, however, suppressed Puma up-regulation and attenuated growth factor withdrawal-induced activation of Bax, DNA fragmentation, and cell death. Conversely, glucose deprivation led to Puma induction even in the presence of growth factor. This regulation of Puma expression was a central component in cell death as a consequence of growth factor or glucose deprivation because Puma deficiency suppressed both of these cell death pathways. Puma induction in growth factor or glucose withdrawal was dependent on p53 in cell lines and in activated primary T lymphocytes because p53 deficiency suppressed Puma induction and delayed Bax and caspase activation, DNA fragmentation, and loss of clonogenic survival. Importantly, although p53 levels did not change or were slightly reduced, p53 activity was suppressed by elevated glucose metabolism to inhibit Puma induction after growth factor withdrawal. These data show that p53 is metabolically regulated and that glucose metabolism initiates a signaling mechanism to inhibit p53 activation and suppress Puma induction, thus promoting an anti-apoptotic balance to Bcl-2 family protein expression that supports cell survival.


Asunto(s)
Glucosa/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Muerte Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Interleucina-3/metabolismo , Ratones , Modelos Biológicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Linfocitos T/metabolismo , Transcripción Genética
10.
Methods Enzymol ; 442: 439-57, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18662583

RESUMEN

Glucose metabolism represents a critical physiological program that not only provides energy to support cell proliferation, but also directly modulates signaling pathways of cell death. With the growing recognition of regulation of cell death by glucose metabolism, many techniques that can be applied in the study have been developed. This chapter discusses several protocols that aid in the analysis of glucose metabolism and cell death and the principles in practicing them under different conditions.


Asunto(s)
Apoptosis/fisiología , Glucosa/metabolismo , Animales , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Muerte Celular/fisiología , Deshidroepiandrosterona/farmacología , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
11.
J Leukoc Biol ; 84(4): 949-57, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18577716

RESUMEN

Lymphocytes require glucose uptake and metabolism for normal survival and function. The signals that regulate the expression and localization of glucose transporter 1 (Glut1) to allow glucose uptake in T cells are now beginning to be understood. Resting T cells require extracellular signals, such as cytokines, hormones, and growth factors, or low-level TCR stimulation to take up adequate glucose to maintain housekeeping functions. In the absence of extrinsic signals, resting T cells internalize and degrade Glut1 and cannot maintain viability. Activated T cells have dramatically increased metabolic requirements to support the energy and biosynthetic needs necessary for growth, proliferation, and effector function. In particular, glucose metabolism and aerobic glycolysis fuel this demand. Therefore, activation of T cells causes a large increase in Glut1 expression and surface localization. If glucose uptake is limited, glycolytic flux decreases to a level that no longer sustains viability, and proapoptotic Bcl-2 family members become activated, promoting cell death. However, excessive glucose uptake can promote hyperactive immune responses and possible immune pathology. Tight regulation of glucose uptake is required to maintain immune homeostasis, and understanding of these metabolic pathways may lead to therapeutic strategies to target some forms of cancer or autoimmunity.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Glucosa/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/prevención & control , Transporte Biológico , Supervivencia Celular , Transportador de Glucosa de Tipo 1/metabolismo , Homeostasis , Humanos , Cinética , Activación de Linfocitos , Linfocitos/citología , Neoplasias/inmunología , Neoplasias/prevención & control , Linfocitos T/citología
12.
J Immunol ; 180(7): 4476-86, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18354169

RESUMEN

T cell activation potently stimulates cellular metabolism to support the elevated energetic and biosynthetic demands of growth, proliferation, and effector function. We show that glucose uptake is limiting in T cell activation and that CD28 costimulation is required to allow maximal glucose uptake following TCR stimulation by up-regulating expression and promoting the cell surface trafficking of the glucose transporter Glut1. Regulation of T cell glucose uptake and Glut1 was critical, as low glucose prevented appropriate T cell responses. Additionally, transgenic expression of Glut1 augmented T cell activation, and led to accumulation of readily activated memory-phenotype T cells with signs of autoimmunity in aged mice. To further examine the regulation of glucose uptake, we analyzed CD28 activation of Akt, which appeared necessary for maximal glucose uptake of stimulated cells and which we have shown can promote Glut1 cell surface trafficking. Consistent with a role for Akt in Glut1 trafficking, transgenic expression of constitutively active myristoylated Akt increased glucose uptake of resting T cells, but did not alter Glut1 protein levels. Therefore, CD28 appeared to promote Akt-independent up-regulation of Glut1 and Akt-dependent Glut1 cell surface trafficking. In support of this model, coexpression of Glut1 and myristoylated Akt transgenes resulted in a synergistic increase in glucose uptake and accumulation of activated T cells in vivo that were largely independent of CD28. Induction of Glut1 protein and Akt regulation of Glut1 trafficking are therefore separable functions of CD28 costimulation that cooperate to promote glucose metabolism for T cell activation and proliferation.


Asunto(s)
Antígenos CD28/metabolismo , Glucosa/metabolismo , Activación de Linfocitos/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Membrana Celular/metabolismo , Tamaño de la Célula , Células Cultivadas , Citocinas/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Regulación de la Expresión Génica , Ratones , Ratones Transgénicos , Transporte de Proteínas , Linfocitos T/citología
13.
Mol Cell Biol ; 27(12): 4328-39, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17371841

RESUMEN

Glucose uptake and utilization are growth factor-stimulated processes that are frequently upregulated in cancer cells and that correlate with enhanced cell survival. The mechanism of metabolic protection from apoptosis, however, has been unclear. Here we identify a novel signaling pathway initiated by glucose catabolism that inhibited apoptotic death of growth factor-deprived cells. We show that increased glucose metabolism protected cells against the proapoptotic Bcl-2 family protein Bim and attenuated degradation of the antiapoptotic Bcl-2 family protein Mcl-1. Maintenance of Mcl-1 was critical for this protection, as glucose metabolism failed to protect Mcl-1-deficient cells from apoptosis. Increased glucose metabolism stabilized Mcl-1 in both cell lines and primary lymphocytes via inhibitory phosphorylation of glycogen synthase kinase 3alpha and 3beta (GSK-3alpha/beta), which otherwise promoted Mcl-1 degradation. While a number of kinases can phosphorylate and inhibit GSK-3alpha/beta, we provide evidence that protein kinase C may be stimulated by glucose-induced alterations in diacylglycerol levels or distribution to phosphorylate GSK-3alpha/beta, maintain Mcl-1 levels, and inhibit cell death. These data provide a novel nutrient-sensitive mechanism linking glucose metabolism and Bcl-2 family proteins via GSK-3 that may promote survival of cells with high rates of glucose utilization, such as growth factor-stimulated or cancerous cells.


Asunto(s)
Apoptosis , Glucosa/metabolismo , Glucógeno Sintasa Quinasa 3/fisiología , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Línea Celular , Glucógeno Sintasa Quinasa 3/clasificación , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA