Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nat Immunol ; 14(5): 489-99, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23563690

RESUMEN

Newly activated CD8(+) T cells reprogram their metabolism to meet the extraordinary biosynthetic demands of clonal expansion; however, the signals that mediate metabolic reprogramming remain poorly defined. Here we demonstrate an essential role for sterol regulatory element-binding proteins (SREBPs) in the acquisition of effector-cell metabolism. Without SREBP signaling, CD8(+) T cells were unable to blast, which resulted in attenuated clonal expansion during viral infection. Mechanistic studies indicated that SREBPs were essential for meeting the heightened lipid requirements of membrane synthesis during blastogenesis. SREBPs were dispensable for homeostatic proliferation, which indicated a context-specific requirement for SREBPs in effector responses. Our studies provide insights into the molecular signals that underlie the metabolic reprogramming of CD8(+) T cells during the transition from quiescence to activation.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Inmunidad Adaptativa/genética , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Transgenes/genética
2.
Proc Natl Acad Sci U S A ; 109(5): 1643-8, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22307624

RESUMEN

Dominant mutations or DNA amplification of tyrosine kinases are rare among the oncogenic alterations implicated in prostate cancer. We demonstrate that castration-resistant prostate cancer (CRPC) in men exhibits increased tyrosine phosphorylation, raising the question of whether enhanced tyrosine kinase activity is observed in prostate cancer in the absence of specific tyrosine kinase mutation or DNA amplification. We generated a mouse model of prostate cancer progression using commonly perturbed non-tyrosine kinase oncogenes and pathways and detected a significant up-regulation of tyrosine phosphorylation at the carcinoma stage. Phosphotyrosine peptide enrichment and quantitative mass spectrometry identified oncogene-specific tyrosine kinase signatures, including activation of EGFR, ephrin type-A receptor 2 (EPHA2), and JAK2. Kinase:substrate relationship analysis of the phosphopeptides also revealed ABL1 and SRC tyrosine kinase activation. The observation of elevated tyrosine kinase signaling in advanced prostate cancer and identification of specific tyrosine kinase pathways from genetically defined tumor models point to unique therapeutic approaches using tyrosine kinase inhibitors for advanced prostate cancer.


Asunto(s)
Oncogenes , Neoplasias de la Próstata/enzimología , Proteínas Tirosina Quinasas/metabolismo , Animales , Activación Enzimática , Masculino , Espectrometría de Masas , Ratones , Fosforilación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteómica , Tirosina/metabolismo , Regulación hacia Arriba
3.
Mol Syst Biol ; 8: 589, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22735335

RESUMEN

The altered metabolism of cancer can render cells dependent on the availability of metabolic substrates for viability. Investigating the signaling mechanisms underlying cell death in cells dependent upon glucose for survival, we demonstrate that glucose withdrawal rapidly induces supra-physiological levels of phospho-tyrosine signaling, even in cells expressing constitutively active tyrosine kinases. Using unbiased mass spectrometry-based phospho-proteomics, we show that glucose withdrawal initiates a unique signature of phospho-tyrosine activation that is associated with focal adhesions. Building upon this observation, we demonstrate that glucose withdrawal activates a positive feedback loop involving generation of reactive oxygen species (ROS) by NADPH oxidase and mitochondria, inhibition of protein tyrosine phosphatases by oxidation, and increased tyrosine kinase signaling. In cells dependent on glucose for survival, glucose withdrawal-induced ROS generation and tyrosine kinase signaling synergize to amplify ROS levels, ultimately resulting in ROS-mediated cell death. Taken together, these findings illustrate the systems-level cross-talk between metabolism and signaling in the maintenance of cancer cell homeostasis.


Asunto(s)
Glucosa/metabolismo , Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/patología , Fosfotirosina/metabolismo , Transducción de Señal/fisiología , Muerte Celular , Línea Celular Tumoral , Retroalimentación Fisiológica , Adhesiones Focales , Humanos , Espectrometría de Masas , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfotirosina/análisis , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
4.
J Proteome Res ; 9(6): 2812-24, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20307106

RESUMEN

Previous studies have shown that oxidized products of the phospholipid PAPC (Ox-PAPC) are strong activators of aortic endothelial cells and play an important role in atherosclerosis and other inflammatory diseases. We and others have demonstrated that Ox-PAPC activates specific signaling pathways and regulates a large number of genes. Using a phosphoproteomic approach based on phosphopeptide enrichment and mass spectrometry analysis, we identified candidate changes in Ox-PAPC-induced protein phosphorylation of 228 proteins. Functional annotation of these proteins showed an enrichment of the regulation of cytoskeleton, junctional components, and tyrosine kinases, all of which may contribute to the phenotypic and molecular changes observed in endothelial cells treated with Ox-PAPC. Many changes in protein phosphorylation induced by Ox-PAPC are reported here for the first time and provide new insights into the mechanism of activation by oxidized lipids, including phosphorylation-based signal transduction.


Asunto(s)
Aorta/citología , Células Endoteliales/metabolismo , Fosfatidilcolinas/metabolismo , Fosfoproteínas/metabolismo , Proteómica/métodos , Secuencia de Aminoácidos , Animales , Aterosclerosis , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Bovinos , Células Cultivadas , Cromatografía por Intercambio Iónico , Quinasas MAP Reguladas por Señal Extracelular/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Datos de Secuencia Molecular , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosfoproteínas/química , Fosforilación , Proteoma/química , Proteoma/metabolismo , Receptor TIE-1/química , Receptor TIE-1/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal
5.
Phys Med Biol ; 51(2): 379-90, 2006 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-16394345

RESUMEN

Many research institutions have a full suite of preclinical tomographic scanners to answer biomedical questions in vivo. Routine multi-modality imaging requires robust registration of images generated by various tomographs. We have implemented a hardware registration method for preclinical imaging that is similar to that used in the combined positron emission tomography (PET)/computed tomography (CT) scanners in the clinic. We designed an imaging chamber which can be rigidly and reproducibly mounted on separate microPET and microCT scanners. We have also designed a three-dimensional grid phantom with 1288 lines that is used to generate the spatial transformation matrix from software registration using a 15-parameter perspective model. The imaging chamber works in combination with the registration phantom synergistically to achieve the image registration goal. We verified that the average registration error between two imaging modalities is 0.335 mm using an in vivo mouse bone scan. This paper also estimates the impact of image misalignment on PET quantitation using attenuation corrections generated from misregistered images. Our technique is expected to produce PET quantitation errors of less than 5%. The methods presented are robust and appropriate for routine use in high throughput animal imaging facilities.


Asunto(s)
Huesos/diagnóstico por imagen , Interpretación de Imagen Asistida por Computador , Fantasmas de Imagen , Tomografía de Emisión de Positrones/instrumentación , Programas Informáticos , Animales , Ratones , Tomografía de Emisión de Positrones/métodos
6.
Cell Discov ; 2: 16028, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27648299

RESUMEN

A prominent mechanism of acquired resistance to BRAF inhibitors in BRAF (V600) -mutant melanoma is associated with the upregulation of receptor tyrosine kinases. Evidences suggested that this resistance mechanism is part of a more complex cellular adaptation process. Using an integrative strategy, we found this mechanism to invoke extensive transcriptomic, (phospho-) proteomic and phenotypic alterations that accompany a cellular transition to a de-differentiated, mesenchymal and invasive state. Even short-term BRAF-inhibitor exposure leads to an early adaptive, differentiation state change-characterized by a slow-cycling, persistent state. The early persistent state is distinct from the late proliferative, resistant state. However, both differentiation states share common signaling alterations including JUN upregulation. Motivated by the similarities, we found that co-targeting of BRAF and JUN is synergistic in killing fully resistant cells; and when used up-front, co-targeting substantially impairs the formation of the persistent subpopulation. We confirmed that JUN upregulation is a common response to BRAF inhibitor treatment in clinically treated patient tumors. Our findings demonstrate that events shared between early- and late-adaptation states provide candidate up-front co-treatment targets.

7.
PLoS One ; 9(1): e84488, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24400094

RESUMEN

Specific oxidized phospholipids (oxPCCD36) promote platelet hyper-reactivity and thrombosis in hyperlipidemia via the scavenger receptor CD36, however the signaling pathway(s) induced in platelets by oxPCCD36 are not well defined. We have employed mass spectrometry-based tyrosine, serine, and threonine phosphoproteomics for the unbiased analysis of platelet signaling pathways induced by oxPCCD36 as well as by the strong physiological agonist thrombin. oxPCCD36 and thrombin induced differential phosphorylation of 115 proteins (162 phosphorylation sites) and 181 proteins (334 phosphorylation sites) respectively. Most of the phosphoproteome changes induced by either agonist have never been reported in platelets; thus they provide candidates in the study of platelet signaling. Bioinformatic analyses of protein phosphorylation dependent responses were used to categorize preferential motifs for (de)phosphorylation, predict pathways and kinase activity, and construct a phosphoproteome network regulating integrin activation. A putative signaling pathway involving Src-family kinases, SYK, and PLCγ2 was identified in platelets activated by oxPCCD36. Subsequent ex vivo studies in human platelets demonstrated that this pathway is downstream of the scavenger receptor CD36 and is critical for platelet activation by oxPCCD36. Our results provide multiple insights into the mechanism of platelet activation and specifically in platelet regulation by oxPCCD36.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Fosfolípidos/farmacología , Fosfoproteínas/metabolismo , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/fisiología , Proteoma , Trombina/farmacología , Antígenos CD36/metabolismo , Análisis por Conglomerados , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfolipasa C gamma/metabolismo , Fosforilación , Mapas de Interacción de Proteínas , Proteínas Tirosina Quinasas/metabolismo , Proteómica , Transducción de Señal/efectos de los fármacos , Quinasa Syk , Trombosis/metabolismo , Familia-src Quinasas/metabolismo
8.
Mol Cancer Res ; 12(12): 1740-54, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25092916

RESUMEN

UNLABELLED: Members of the Ewing sarcoma family of tumors (ESFT) contain tumor-associated translocations that give rise to oncogenic transcription factors, most commonly EWS/FLI1. EWS/FLI1 plays a dominant role in tumor progression by modulating the expression of hundreds of target genes. Here, the impact of EWS/FLI1 inhibition, by RNAi-mediated knockdown, on cellular signaling was investigated using mass spectrometry-based phosphoproteomics to quantify global changes in phosphorylation. This unbiased approach identified hundreds of unique phosphopeptides enriched in processes such as regulation of cell cycle and cytoskeleton organization. In particular, phosphotyrosine profiling revealed a large upregulation of STAT3 phosphorylation upon EWS/FLI1 knockdown. However, single-cell analysis demonstrated that this was not a cell-autonomous effect of EWS/FLI1 deficiency, but rather a signaling effect occurring in cells in which knockdown does not occur. Conditioned media from knockdown cells were sufficient to induce STAT3 phosphorylation in control cells, verifying the presence of a soluble factor that can activate STAT3. Cytokine analysis and ligand/receptor inhibition experiments determined that this activation occurred, in part, through an IL6-dependent mechanism. Taken together, the data support a model in which EWS/FLI1 deficiency results in the secretion of soluble factors, such as IL6, which activate STAT signaling in bystander cells that maintain EWS/FLI1 expression. Furthermore, these soluble factors were shown to protect against apoptosis. IMPLICATIONS: EWS/FLI1 inhibition results in a novel adaptive response and suggests that targeting the IL6/STAT3 signaling pathway may increase the efficacy of ESFT therapies.


Asunto(s)
Neoplasias Óseas/metabolismo , Interleucina-6/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteómica/métodos , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Factor de Transcripción STAT3/metabolismo , Sarcoma de Ewing/metabolismo , Apoptosis , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Comunicación Paracrina , Fosforilación
9.
Cell Metab ; 19(4): 694-701, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24703700

RESUMEN

Virus infections trigger metabolic changes in host cells that support the bioenergetic and biosynthetic demands of viral replication. Although recent studies have characterized virus-induced changes in host cell metabolism (Munger et al., 2008; Terry et al., 2012), the molecular mechanisms by which viruses reprogram cellular metabolism have remained elusive. Here, we show that the gene product of adenovirus E4ORF1 is necessary for adenovirus-induced upregulation of host cell glucose metabolism and sufficient to promote enhanced glycolysis in cultured epithelial cells by activation of MYC. E4ORF1 localizes to the nucleus, binds to MYC, and enhances MYC binding to glycolytic target genes, resulting in elevated expression of specific glycolytic enzymes. E4ORF1 activation of MYC promotes increased nucleotide biosynthesis from glucose intermediates and enables optimal adenovirus replication in primary lung epithelial cells. Our findings show how a viral protein exploits host cell machinery to reprogram cellular metabolism and promote optimal progeny virion generation.


Asunto(s)
Proteínas E4 de Adenovirus/metabolismo , Células Epiteliales/metabolismo , Glucosa/metabolismo , Redes y Vías Metabólicas/fisiología , Modelos Biológicos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Replicación Viral/fisiología , Proteínas E4 de Adenovirus/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Redes y Vías Metabólicas/genética , Nucleótidos/biosíntesis , Unión Proteica , Replicación Viral/genética
10.
Science ; 339(6126): 1448-53, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23449998

RESUMEN

Type I interferons (IFN-α and IFN-ß) are important for protection against many viral infections, whereas type II interferon (IFN-γ) is essential for host defense against some bacterial and parasitic pathogens. Study of IFN responses in human leprosy revealed an inverse correlation between IFN-ß and IFN-γ gene expression programs. IFN-γ and its downstream vitamin D-dependent antimicrobial genes were preferentially expressed in self-healing tuberculoid lesions and mediated antimicrobial activity against the pathogen Mycobacterium leprae in vitro. In contrast, IFN-ß and its downstream genes, including interleukin-10 (IL-10), were induced in monocytes by M. leprae in vitro and preferentially expressed in disseminated and progressive lepromatous lesions. The IFN-γ-induced macrophage vitamin D-dependent antimicrobial peptide response was inhibited by IFN-ß and by IL-10, suggesting that the differential production of IFNs contributes to protection versus pathogenesis in some human bacterial infections.


Asunto(s)
Interferón beta/inmunología , Interferón gamma/inmunología , Lepra Lepromatosa/inmunología , Lepra Tuberculoide/inmunología , Mycobacterium leprae/inmunología , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Humanos , Interferón beta/genética , Interferón beta/metabolismo , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Lepra Lepromatosa/genética , Lepra Lepromatosa/metabolismo , Lepra Tuberculoide/genética , Lepra Tuberculoide/metabolismo , Viabilidad Microbiana , Monocitos/inmunología , Monocitos/metabolismo , Mycobacterium leprae/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Transcriptoma , Tuberculosis/genética , Tuberculosis/inmunología , Regulación hacia Arriba , beta-Defensinas/genética , beta-Defensinas/metabolismo , Catelicidinas
11.
Cancer Res ; 73(9): 2850-62, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23440422

RESUMEN

The sterol regulatory element-binding proteins (SREBP) are key transcriptional regulators of lipid metabolism and cellular growth. It has been proposed that SREBP signaling regulates cellular growth through its ability to drive lipid biosynthesis. Unexpectedly, we find that loss of SREBP activity inhibits cancer cell growth and viability by uncoupling fatty acid synthesis from desaturation. Integrated lipid profiling and metabolic flux analysis revealed that cancer cells with attenuated SREBP activity maintain long-chain saturated fatty acid synthesis, while losing fatty acid desaturation capacity. We traced this defect to the uncoupling of fatty acid synthase activity from stearoyl-CoA desaturase 1 (SCD1)-mediated desaturation. This deficiency in desaturation drives an imbalance between the saturated and monounsaturated fatty acid pools resulting in severe lipotoxicity. Importantly, replenishing the monounsaturated fatty acid pool restored growth to SREBP-inhibited cells. These studies highlight the importance of fatty acid desaturation in cancer growth and provide a novel mechanistic explanation for the role of SREBPs in cancer metabolism.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias/metabolismo , Animales , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Ácido Graso Sintasas/metabolismo , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos NOD , Modelos Estadísticos , Trasplante de Neoplasias , Transducción de Señal , Estearoil-CoA Desaturasa/metabolismo , Esteroles/metabolismo
12.
Science ; 340(6132): 626-30, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23558169

RESUMEN

The recent discovery of mutations in metabolic enzymes has rekindled interest in harnessing the altered metabolism of cancer cells for cancer therapy. One potential drug target is isocitrate dehydrogenase 1 (IDH1), which is mutated in multiple human cancers. Here, we examine the role of mutant IDH1 in fully transformed cells with endogenous IDH1 mutations. A selective R132H-IDH1 inhibitor (AGI-5198) identified through a high-throughput screen blocked, in a dose-dependent manner, the ability of the mutant enzyme (mIDH1) to produce R-2-hydroxyglutarate (R-2HG). Under conditions of near-complete R-2HG inhibition, the mIDH1 inhibitor induced demethylation of histone H3K9me3 and expression of genes associated with gliogenic differentiation. Blockade of mIDH1 impaired the growth of IDH1-mutant--but not IDH1-wild-type--glioma cells without appreciable changes in genome-wide DNA methylation. These data suggest that mIDH1 may promote glioma growth through mechanisms beyond its well-characterized epigenetic effects.


Asunto(s)
Bencenoacetamidas/farmacología , Diferenciación Celular , Inhibidores Enzimáticos/farmacología , Glioma/enzimología , Glioma/patología , Imidazoles/farmacología , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Animales , Bencenoacetamidas/administración & dosificación , Bencenoacetamidas/toxicidad , Diferenciación Celular/efectos de los fármacos , Transformación Celular Neoplásica , Inhibidores Enzimáticos/toxicidad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Glioma/genética , Glutaratos/metabolismo , Histonas/metabolismo , Imidazoles/administración & dosificación , Imidazoles/toxicidad , Isocitrato Deshidrogenasa/química , Isocitrato Deshidrogenasa/metabolismo , Metilación , Ratones , Ratones SCID , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Multimerización de Proteína , Interferencia de ARN , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Nat Med ; 18(4): 555-63, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22447076

RESUMEN

It is unclear whether the ability of the innate immune system to recognize distinct ligands from a single microbial pathogen via multiple pattern recognition receptors (PRRs) triggers common pathways or differentially triggers specific host responses. In the human mycobacterial infection leprosy, we found that activation of monocytes via nucleotide-binding oligomerization domain-containing protein 2 (NOD2) by its ligand muramyl dipeptide, as compared to activation via heterodimeric Toll-like receptor 2 and Toll-like receptor 1 (TLR2/1) by triacylated lipopeptide, preferentially induced differentiation into dendritic cells (DCs), which was dependent on a previously unknown interleukin-32 (IL-32)-dependent mechanism. Notably, IL-32 was sufficient to induce monocytes to rapidly differentiate into DCs, which were more efficient than granulocyte-macrophage colony-stimulating factor (GM-CSF)-derived DCs in presenting antigen to major histocompatibility complex (MHC) class I-restricted CD8(+) T cells. Expression of NOD2 and IL-32 and the frequency of CD1b(+) DCs at the site of leprosy infection correlated with the clinical presentation; they were greater in patients with limited as compared to progressive disease. The addition of recombinant IL-32 restored NOD2-induced DC differentiation in patients with the progressive form of leprosy. In conclusion, the NOD2 ligand-induced, IL-32-dependent DC differentiation pathway contributes a key and specific mechanism for host defense against microbial infection in humans.


Asunto(s)
Células Dendríticas/metabolismo , Interleucinas/metabolismo , Lepra/patología , Proteína Adaptadora de Señalización NOD2/metabolismo , Antígenos CD , Antígeno CD11b , Diferenciación Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucinas/farmacología , Ligandos , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Componente Principal , ARN Mensajero/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
14.
Nat Med ; 18(2): 267-73, 2012 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-22286305

RESUMEN

Leprosy provides a model to investigate mechanisms of immune regulation in humans, given that the disease forms a spectrum of clinical presentations that correlate with host immune responses. Here we identified 13 miRNAs that were differentially expressed in the lesions of subjects with progressive lepromatous (L-lep) versus the self-limited tuberculoid (T-lep) disease. Bioinformatic analysis revealed a significant enrichment of L-lep-specific miRNAs that preferentially target key immune genes downregulated in L-lep versus T-lep lesions. The most differentially expressed miRNA in L-lep lesions, hsa-mir-21, was upregulated in Mycobacterium leprae-infected monocytes. By directly downregulating Toll-like receptor 2/1 heterodimer (TLR2/1)-induced CYP27B1 and IL1B expression as well as indirectly upregulating interleukin-10 (IL-10), hsa-mir-21 inhibited expression of the genes encoding two vitamin D-dependent antimicrobial peptides, CAMP and DEFB4A. Conversely, knockdown of hsa-mir-21 in M. leprae-infected monocytes enhanced expression of CAMP and DEFB4A and restored TLR2/1-mediated antimicrobial activity against M. leprae. Therefore, the ability of M. leprae to upregulate hsa-mir-21 targets multiple genes associated with the immunologically localized disease form, providing an effective mechanism to escape from the vitamin D-dependent antimicrobial pathway.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Lepra/inmunología , MicroARNs/fisiología , Vitamina D/fisiología , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/fisiología , Células Cultivadas , Humanos , Interleucina-10/fisiología , Interleucina-1beta/fisiología , Lepra Lepromatosa/inmunología , Lepra Tuberculoide/inmunología , MicroARNs/inmunología , Monocitos/inmunología , Monocitos/microbiología , Mycobacterium leprae/inmunología , Transducción de Señal/fisiología , Receptor Toll-Like 1/inmunología , Receptor Toll-Like 2/inmunología , beta-Defensinas/fisiología
15.
Sci Signal ; 4(166): ra18, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21447799

RESUMEN

In subtypes and late stages of leukemias driven by the tyrosine kinase fusion protein Bcr-Abl, signaling by the Src family kinases (SFKs) critically contributes to the leukemic phenotype. We performed global tyrosine phosphoprofiling by quantitative mass spectrometry of Bcr-Abl-transformed cells in which the activities of the SFKs were perturbed to build a detailed context-dependent network of cancer signaling. Perturbation of the SFKs Lyn and Hck with genetics or inhibitors revealed Bcr-Abl downstream phosphorylation events either mediated by or independent of SFKs. We identified multiple negative feedback mechanisms within the network of signaling events affected by Bcr-Abl and SFKs and found that Bcr-Abl attenuated these inhibitory mechanisms. The C-terminal Src kinase (Csk)-binding protein Pag1 (also known as Cbp) and the tyrosine phosphatase Ptpn18 both mediated negative feedback to SFKs. We observed Bcr-Abl-mediated phosphorylation of the phosphatase Shp2 (Ptpn11), and this may contribute to the suppression of these negative feedback mechanisms to promote Bcr-Abl-activated SFK signaling. Csk and a kinase-deficient Csk mutant both produced similar globally repressive signaling consequences, suggesting a critical role for the adaptor protein function of Csk in its inhibition of Bcr-Abl and SFK signaling. The identified Bcr-Abl-activated SFK regulatory mechanisms are candidates for dysregulation during leukemia progression and acquisition of SFK-mediated drug resistance.


Asunto(s)
Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/metabolismo , Leucemia/enzimología , Proteínas Tirosina Quinasas/metabolismo , Proteómica , Transducción de Señal , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Tirosina Quinasa CSK , Línea Celular Transformada , Línea Celular Tumoral , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia/tratamiento farmacológico , Leucemia/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fosforilación , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
16.
Cancer Res ; 71(15): 5164-74, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21646475

RESUMEN

In contrast to normal cells, cancer cells avidly take up glucose and metabolize it to lactate even when oxygen is abundant, a phenomenon referred to as the Warburg effect. This fundamental alteration in glucose metabolism in cancer cells enables their specific detection by positron emission tomography (PET) following i.v. injection of the glucose analogue (18)F-fluorodeoxy-glucose ((18)FDG). However, this useful imaging technique is limited by the fact that not all cancers avidly take up FDG. To identify molecular determinants of (18)FDG retention, we interrogated the transcriptomes of human-cancer cell lines and primary tumors for metabolic pathways associated with (18)FDG radiotracer uptake. From ninety-five metabolic pathways that were interrogated, the glycolysis, and several glycolysis-related pathways (pentose phosphate, carbon fixation, aminoacyl-tRNA biosynthesis, one-carbon-pool by folate) showed the greatest transcriptional enrichment. This "FDG signature" predicted FDG uptake in breast cancer cell lines and overlapped with established gene expression signatures for the "basal-like" breast cancer subtype and MYC-induced tumorigenesis in mice. Human breast cancers with nuclear MYC staining and high RNA expression of MYC target genes showed high (18)FDG-PET uptake (P < 0.005). Presence of the FDG signature was similarly associated with MYC gene copy gain, increased MYC transcript levels, and elevated expression of metabolic MYC target genes in a human breast cancer genomic dataset. Together, our findings link clinical observations of glucose uptake with a pathologic and molecular subtype of human breast cancer. Furthermore, they suggest related approaches to derive molecular determinants of radiotracer retention for other PET-imaging probes.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias de la Mama/metabolismo , Radioisótopos de Flúor , Fluorodesoxiglucosa F18 , Perfilación de la Expresión Génica , Genes myc , Glucólisis , Proteínas de Neoplasias/biosíntesis , Tomografía de Emisión de Positrones , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Radiofármacos , Adenocarcinoma/clasificación , Adenocarcinoma/patología , Astrocitoma/metabolismo , Astrocitoma/patología , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/patología , Línea Celular Tumoral/metabolismo , Femenino , Radioisótopos de Flúor/farmacocinética , Fluorodesoxiglucosa F18/farmacocinética , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Glucólisis/genética , Humanos , Masculino , Melanoma/patología , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/patología , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Radiofármacos/farmacocinética
17.
Proc Natl Acad Sci U S A ; 103(51): 19466-71, 2006 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-17164333

RESUMEN

The success of targeting kinases in cancer with small molecule inhibitors has been tempered by the emergence of drug-resistant kinase domain mutations. In patients with chronic myeloid leukemia treated with ABL inhibitors, BCR-ABL kinase domain mutations are the principal mechanism of relapse. Certain mutations are occasionally detected before treatment, suggesting increased fitness relative to wild-type p210 BCR-ABL. We evaluated the oncogenicity of eight kinase inhibitor-resistant BCR-ABL mutants and found a spectrum of potencies greater or less than p210. Although most fitness alterations correlate with changes in kinase activity, this is not the case with the T315I BCR-ABL mutation that confers clinical resistance to all currently approved ABL kinase inhibitors. Through global phosphoproteome analysis, we identified a unique phosphosubstrate signature associated with each drug-resistant allele, including a shift in phosphorylation of two tyrosines (Tyr253 and Tyr257) in the ATP binding loop (P-loop) of BCR-ABL when Thr315 is Ile or Ala. Mutational analysis of these tyrosines in the context of Thr315 mutations demonstrates that the identity of the gatekeeper residue impacts oncogenicity by altered P-loop phosphorylation. Therefore, mutations that confer clinical resistance to kinase inhibitors can substantially alter kinase function and confer novel biological properties that may impact disease progression.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Análisis Mutacional de ADN , Espectrometría de Masas , Ratones , Datos de Secuencia Molecular , Mutación Missense/genética , Fosforilación , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Proteómica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA